Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
BMC Biol ; 17(1): 56, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31311534

RESUMEN

BACKGROUND: Adaptive responses to stress are essential for cell and organismal survival. In metazoans, little is known about the impact of environmental stress on RNA homeostasis. RESULTS: By studying the regulation of a cadmium-induced gene named numr-1 in Caenorhabditis elegans, we discovered that disruption of RNA processing acts as a signal for environmental stress. We find that NUMR-1 contains motifs common to RNA splicing factors and influences RNA splicing in vivo. A genome-wide screen reveals that numr-1 is strongly and specifically induced by silencing of genes that function in basal RNA metabolism including subunits of the metazoan integrator complex. Human integrator processes snRNAs for functioning with splicing factors, and we find that silencing of C. elegans integrator subunits disrupts snRNA processing, causes aberrant pre-mRNA splicing, and induces the heat shock response. Cadmium, which also strongly induces numr-1, has similar effects on RNA and the heat shock response. Lastly, we find that heat shock factor-1 is required for full numr-1 induction by cadmium. CONCLUSION: Our results are consistent with a model in which disruption of integrator processing of RNA acts as a molecular damage signal initiating an adaptive stress response mediated by heat shock factor-1. When numr-1 is induced via this pathway in C. elegans, its function in RNA metabolism may allow it to mitigate further damage and thereby promote tolerance to cadmium.


Asunto(s)
Cadmio/toxicidad , Caenorhabditis elegans/fisiología , Regulación de la Expresión Génica , Respuesta al Choque Térmico/fisiología , Procesamiento Postranscripcional del ARN/fisiología , Empalme del ARN , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Respuesta al Choque Térmico/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Estrés Fisiológico
2.
PLoS Genet ; 12(10): e1006361, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27776126

RESUMEN

SKN-1/Nrf are the primary antioxidant/detoxification response transcription factors in animals and they promote health and longevity in many contexts. SKN-1/Nrf are activated by a remarkably broad-range of natural and synthetic compounds and physiological conditions. Defining the signaling mechanisms that regulate SKN-1/Nrf activation provides insights into how cells coordinate responses to stress. Nrf2 in mammals is regulated in part by the redox sensor repressor protein named Keap1. In C. elegans, the p38 MAPK cascade in the intestine activates SKN-1 during oxidative stress by promoting its nuclear accumulation. Interestingly, we find variation in the kinetics of p38 MAPK activation and tissues with SKN-1 nuclear accumulation among different pro-oxidants that all trigger strong induction of SKN-1 target genes. Using genome-wide RNAi screening, we identify new genes that are required for activation of the core SKN-1 target gene gst-4 during exposure to the natural pro-oxidant juglone. Among 10 putative activators identified in this screen was skr-1/2, highly conserved homologs of yeast and mammalian Skp1, which function to assemble protein complexes. Silencing of skr-1/2 inhibits induction of SKN-1 dependent detoxification genes and reduces resistance to pro-oxidants without decreasing p38 MAPK activation. Global transcriptomics revealed strong correlation between genes that are regulated by SKR-1/2 and SKN-1 indicating a high degree of specificity. We also show that SKR-1/2 functions upstream of the WD40 repeat protein WDR-23, which binds to and inhibits SKN-1. Together, these results identify a novel p38 MAPK independent signaling mechanism that activates SKN-1 via SKR-1/2 and involves WDR-23.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Inactivación Metabólica/genética , Longevidad/genética , Proteínas Ligasas SKP Cullina F-box/genética , Receptores de Activinas Tipo I/genética , Animales , Antioxidantes/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/biosíntesis , Tracto Gastrointestinal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/biosíntesis , Proteína 1 Asociada A ECH Tipo Kelch/genética , Fosforilación , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Ligasas SKP Cullina F-box/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
J Exp Biol ; 219(Pt 14): 2201-11, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27207646

RESUMEN

It has long been recognized that simultaneous exposure to heat stress and oxidative stress shows a synergistic interaction that reduces organismal fitness, but relatively little is known about the mechanisms underlying this interaction. We investigated the role of molecular stress responses in driving this synergistic interaction using the nematode Caenorhabditis elegans To induce oxidative stress, we used the pro-oxidant compounds acrylamide, paraquat and juglone. As expected, we found that heat stress and oxidative stress interact synergistically to reduce survival. Compared with exposure to each stressor alone, during simultaneous sublethal exposure to heat stress and oxidative stress the normal induction of key oxidative-stress response (OxSR) genes was generally inhibited, whereas the induction of key heat-shock response (HSR) genes was not. Genetically activating the SKN-1-dependent OxSR increased a marker for protein aggregation and decreased whole-worm survival during heat stress alone, with the latter being independent of HSF-1. In contrast, compared with wild-type worms, inactivating the HSR by HSF-1 knockdown, which would be expected to decrease basal heat shock protein expression, increased survival during oxidative stress alone. Taken together, these data suggest that, in C. elegans, the HSR and OxSR cannot be simultaneously activated to the same extent that each can be activated during a single stressor exposure. We conclude that the observed synergistic reduction in survival during combined exposure to heat stress and oxidative stress is due, at least in part, to inhibition of the OxSR during activation of the HSR.


Asunto(s)
Caenorhabditis elegans/fisiología , Respuesta al Choque Térmico/fisiología , Estrés Oxidativo , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/genética , Animales , Biomarcadores/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Ambiente , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Respuesta al Choque Térmico/genética , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Agregado de Proteínas/efectos de los fármacos , Interferencia de ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Análisis de Supervivencia , Factores de Transcripción/metabolismo
4.
ACS Chem Biol ; 19(5): 1180-1193, 2024 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-38652683

RESUMEN

C. elegans numr-1/2 (nuclear-localized metal-responsive) is an identical gene pair encoding a nuclear protein previously shown to be activated by cadmium and disruption of the integrator RNA metabolism complex. We took a chemical genetic approach to further characterize regulation of this novel metal response by screening 41,716 compounds and extracts for numr-1p::GFP activation. The most potent activator was chaetocin, a fungal 3,6-epidithiodiketopiperazine (ETP) with promising anticancer activity. Chaetocin activates numr-1/2 strongly in the alimentary canal but is distinct from metal exposure, because it represses canonical cadmium-responsive metallothionine genes. Chaetocin has diverse targets in cancer cells including thioredoxin reductase, histone lysine methyltransferase, and acetyltransferase p300/CBP; further work is needed to identify the mechanism in C. elegans as genetic disruption and RNAi screening of homologues did not induce numr-1/2 in the alimentary canal and chaetocin did not affect markers of integrator dysfunction. We demonstrate that disulfides in chaetocin and chetomin, a dimeric ETP analog, are required to induce numr-1/2. ETP monomer gliotoxin, despite possessing a disulfide linkage, had almost no effect on numr-1/2, suggesting a dimer requirement. Chetomin inhibits C. elegans growth at low micromolar levels, and loss of numr-1/2 increases sensitivity; C. elegans and Chaetomiaceae fungi inhabit similar environments raising the possibility that numr-1/2 functions as a defense mechanism. There is no direct orthologue of numr-1/2 in humans, but RNaseq suggests that chaetocin affects expression of cellular processes linked to stress response and metal homeostasis in colorectal cancer cells. Our results reveal interactions between metal response gene regulation and ETPs and identify a potential mechanism of resistance to this versatile class of preclinical compounds.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Homeostasis , Micotoxinas , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Micotoxinas/farmacología , Micotoxinas/metabolismo , Homeostasis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Piperazinas/farmacología , Piperazinas/química , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Cadmio/farmacología
5.
Am J Physiol Regul Integr Comp Physiol ; 305(3): R175-86, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23739341

RESUMEN

Intracellular salt and water homeostasis is essential for all cellular life. Extracellular salt and water homeostasis is also important for multicellular organisms. Many fundamental mechanisms of compensation for osmotic perturbations are well defined and conserved. Alternatively, molecular mechanisms of detecting salt and water imbalances and regulating compensatory responses are generally poorly defined for animals. Throughout the last century, researchers studying vertebrates and vertebrate cells made critical contributions to our understanding of osmoregulation, especially mechanisms of salt and water transport and organic osmolyte accumulation. Researchers have more recently started using invertebrate model organisms with defined genomes and well-established methods of genetic manipulation to begin defining the genes and integrated regulatory networks that respond to osmotic stress. The nematode Caenorhabditis elegans is well suited to these studies. Here, I introduce osmoregulatory mechanisms in this model, discuss experimental advantages and limitations, and review important findings. Key discoveries include defining genetic mechanisms of osmolarity sensing in neurons, identifying protein damage as a sensor and principle determinant of hypertonic stress resistance, and identification of a putative sensor for hypertonic stress associated with the extracellular matrix. Many of these processes and pathways are conserved and, therefore, provide new insights into salt and water homeostasis in other animals, including mammals.


Asunto(s)
Caenorhabditis elegans/fisiología , Electrólitos/metabolismo , Homeostasis/fisiología , Equilibrio Hidroelectrolítico/fisiología , Agua/metabolismo , Animales , Conducta Animal/fisiología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Soluciones Hipertónicas , Transducción de Señal/fisiología
6.
Am J Physiol Regul Integr Comp Physiol ; 305(11): R1376-89, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24049119

RESUMEN

Ca(2+)-activated Cl(-) channels (CaCCs) are critical to processes such as epithelial transport, membrane excitability, and signal transduction. Anoctamin, or TMEM16, is a family of 10 mammalian transmembrane proteins, 2 of which were recently shown to function as CaCCs. The functions of other family members have not been firmly established, and almost nothing is known about anoctamins in invertebrates. Therefore, we performed a phylogenetic analysis of anoctamins across the animal kingdom and examined the expression and function of anoctamins in the genetically tractable nematode Caenorhabditis elegans. Phylogenetic analyses support five anoctamin clades that are at least as old as the deuterostome/protosome ancestor. This includes a branch containing two Drosophila paralogs that group with mammalian ANO1 and ANO2, the two best characterized CaCCs. We identify two anoctamins in C. elegans (ANOH-1 and ANOH-2) that are also present in basal metazoans. The anoh-1 promoter is active in amphid sensory neurons that detect external chemical and nociceptive cues. Within amphid neurons, ANOH-1::GFP fusion protein is enriched within sensory cilia. RNA interference silencing of anoh-1 reduced avoidance of steep osmotic gradients without disrupting amphid cilia development, chemotaxis, or withdrawal from noxious stimuli, suggesting that ANOH-1 functions in a sensory mode-specific manner. The anoh-2 promoter is active in mechanoreceptive neurons and the spermatheca, but loss of anoh-2 had no effect on motility or brood size. Our study indicates that at least five anoctamin duplicates are evolutionarily ancient and suggests that sensory signaling may be a basal function of the anoctamin protein family.


Asunto(s)
Caenorhabditis elegans/metabolismo , Canales de Cloruro/metabolismo , Proteínas de la Membrana/metabolismo , Filogenia , Animales , Transporte Biológico/genética , Caenorhabditis elegans/genética , Calcio/metabolismo , Canales de Cloruro/genética , Proteínas de la Membrana/genética , Neuronas Aferentes/metabolismo , Transducción de Señal/genética
7.
PLoS One ; 18(5): e0285328, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37155688

RESUMEN

In epidermal tissues, extracellular matrices (ECMs) function as barriers between the organism and environment. Despite being at the interface with the environment, little is known about the role of animal barrier ECMs in sensing stress and communicating with cytoprotective gene pathways in neighboring cells. We and others have identified a putative damage sensor in the C. elegans cuticle that regulates osmotic, detoxification, and innate immune response genes. This pathway is associated with circumferential collagen bands called annular furrows; mutation or loss of furrow collagens causes constitutive activation of osmotic, detoxification, and innate immune response genes. Here, we performed a genome-wide RNAi screen for modulators of osmotic stress response gene gpdh-1 in a furrow collagen mutant strain. RNAi of six genes identified in this screen were tested under other conditions and for effects on other stress responses. The functions of these genes suggest negative feedback within osmolyte accumulation pathways and interactions with ATP homeostasis and protein synthesis. Loss of these gpdh-1 modulators had distinct effects on canonical detoxification and innate immune response genes.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Interferencia de ARN , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Retroalimentación , Matriz Extracelular/metabolismo , Colágeno/metabolismo
8.
Aging Cell ; 22(4): e13795, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36797658

RESUMEN

CCR4-NOT is a versatile eukaryotic protein complex that controls multiple steps in gene expression regulation from synthesis to decay. In yeast, CCR4-NOT has been implicated in stress response regulation, though this function in other organisms remains unclear. In a genome-wide RNAi screen, we identified a subunit of the CCR4-NOT complex, ccf-1, as a requirement for the C. elegans transcriptional response to cadmium and acrylamide stress. Using whole-transcriptome RNA sequencing, we show that the knockdown of ccf-1 attenuates the activation of a broad range of stress-protective genes in response to cadmium and acrylamide, including those encoding heat shock proteins and xenobiotic detoxification. Consistently, survival assays show that the knockdown of ccf-1 decreases C. elegans stress resistance and normal lifespan. A yeast 2-hybrid screen using a CCF-1 bait identified the homeobox transcription factor PAL-1 as a physical interactor. Knockdown of pal-1 inhibits the activation of ccf-1 dependent stress genes and reduces C. elegans stress resistance. Gene expression analysis reveals that knockdown of ccf-1 and pal-1 attenuates the activation of elt-2 and elt-3 under stress that encode master transcriptional co-regulators of stress response in the C. elegans, and that overexpression of ELT-2 can suppress ccf-1's requirement for gene transcription in a stress-dependent manner. Our findings reveal a new role for CCR4-NOT in the environmental stress response and define its role in stress resistance and longevity in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans , Proteínas de Saccharomyces cerevisiae , Animales , Acrilamidas , Cadmio/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Longevidad/genética , Ribonucleasas/genética , Ribonucleasas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
9.
Drug Metab Rev ; 44(3): 209-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22656429

RESUMEN

Nematodes parasitize an alarming number of people and agricultural animals globally and cause debilitating morbidity and mortality. Anthelmintics have been the primary tools used to control parasitic nematodes for the past several decades, but drug resistance is becoming a major obstacle. Xenobiotic detoxification pathways defend against drugs and other foreign chemicals in diverse organisms, and evidence is accumulating that they play a role in mediating resistance to anthelmintics in nematodes. Related antioxidation pathways may also provide filarial parasites with protection against host free-radical-mediated immune responses. Upstream regulatory pathways have received almost no attention in nematode parasites, despite their potential to coregulate multiple detoxification and antioxidation genes. The nuclear eurythroid 2-related factor 2 (NRF2) transcription factor mediates inducible detoxification and antioxidation defenses in mammals, and recent studies have demonstrated that it promotes multidrug resistance in some human tumors. Recent studies in the free-living model nematode, Caenorhabditis elegans, have defined the homologous transcription factor, SKN-1, as a master regulator of detoxification and antioxidation genes. Despite similar functions, SKN-1 and NRF2 have important differences in structure and regulatory pathways. Protein alignment and phylogenetic analyses indicate that these differences are shared among many nematodes, making SKN-1 a candidate for specifically targeting nematode detoxification and antioxidation.


Asunto(s)
Antihelmínticos/farmacología , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Resistencia a Medicamentos , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Antihelmínticos/farmacocinética , Proteínas de Caenorhabditis elegans/química , Proteínas de Unión al ADN/química , Resistencia a Múltiples Medicamentos , Proteínas del Helminto/química , Proteínas del Helminto/metabolismo , Humanos , Inactivación Metabólica , Datos de Secuencia Molecular , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/metabolismo , Nematodos/efectos de los fármacos , Nematodos/metabolismo , Filogenia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factores de Transcripción/química
10.
Genetics ; 220(3)2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34849856

RESUMEN

We and others previously identified circumferential bands of collagen named annular furrows as key components of a damage sensor in the cuticle of Caenorhabditis elegans that regulates cytoprotective genes. Mutation or loss of noncollagen secreted proteins OSM-7, OSM-8, and OSM-11 activate the same cytoprotective responses without obvious changes to the cuticle indicating that other extracellular proteins are involved. Here, we used RNAi screening to identify protein kinase DRL-1 as a key modulator of cytoprotective gene expression and stress resistance in furrow and extracellular OSM protein mutants. DRL-1 functions downstream from furrow disruption and is expressed in cells that induce cytoprotective genes. DRL-1 is not required for the expression of cytoprotective genes under basal or oxidative stress conditions consistent with specificity to extracellular signals. DRL-1 was previously shown to regulate longevity via a "Dietary Restriction-Like" state, but it functions downstream from furrow disruption by a distinct mechanism. The kinase domain of DRL-1 is related to mammalian MEKK3, and MEKK3 is recruited to a plasma membrane osmosensor complex by a scaffold protein. In C. elegans, DRL-1 contains an atypical hydrophobic C-terminus with predicted transmembrane domains and is constitutively expressed at or near the plasma membrane where it could function to receive extracellular damage signals for cells that mount cytoprotective responses.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Matriz Extracelular/metabolismo , Longevidad/genética , Mamíferos , Interferencia de ARN
11.
G3 (Bethesda) ; 12(11)2022 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-36000892

RESUMEN

Mutation or loss of 6 extracellular matrix collagen genes disrupts annular furrows in adult C. elegans cuticles, causes a wide "Dumpy" body morphology, and activates osmotic, detoxification, and antimicrobial defense genes. High environmental osmolarity reduces internal turgor pressure, physically distorts the epidermis, and activates the same stress responses. Collagen gene mutations that cause Dumpy without furrow disruption do not activate stress responses. These results are consistent with an extracellular damage sensor associated with furrows in the adult cuticle that regulates environmental stress responses in adjacent cells. Several cuticle characteristics change between molts, but all stages have annular furrows and express furrow collagen genes. We compared body shape, furrow organization imaged with differential interference contrast microscopy, and stress response gene expression in furrow collagen gene mutants at all postembryonic stages. We find that most body shape and furrow disorganization phenotypes start at the L3 stage and increase in severity with each molt afterwards. Stress response genes were induced the strongest in adults, correlating with the greatest Dumpy and furrow phenotypes. Although weaker than in adults, osmolyte transporter gene hmit-1.1 and antimicrobial gene nlp-29 were also induced in some early larvae that had weak or undetectable cuticle phenotypes. Our data are consistent with progressive cuticle phenotypes in which each new cuticle is at least partially directed by organization of the former cuticle. Gene expression and cuticle data support the role of furrow disruption as a signal in L4 larvae and adults, but also suggest a role for other cuticle organization or epidermal cell effects in early larvae.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Colágeno/genética , Fenotipo , Larva/genética , Larva/metabolismo
12.
PLoS One ; 17(8): e0272452, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35951614

RESUMEN

Physiological responses to the environment, disease, and aging vary by sex in many animals, but mechanisms of dimorphism have only recently begun to receive careful attention. The genetic model nematode Caenorhabditis elegans has well-defined mechanisms of stress response, aging, and sexual differentiation. C. elegans has males, but the vast majority of research only uses hermaphrodites. We found that males of the standard N2 laboratory strain were more resistant to hyperosmolarity, heat, and a natural pro-oxidant than hermaphrodites when in mixed-sex groups. Resistance to heat and pro-oxidant were also male-biased in three genetically and geographically diverse C. elegans strains consistent with a species-wide dimorphism that is not specific to domestication. N2 males were also more resistant to heat and pro-oxidant when keep individually indicating that differences in resistance do not require interactions between worms. We found that males induce canonical stress response genes by similar degrees and in similar tissues as hermaphrodites suggesting the importance of other mechanisms. We find that resistance to heat and pro-oxidant are influenced by the sex differentiation transcription factor TRA-1 suggesting that downstream organ differentiation pathways establish differences in stress resistance. Environmental stress influences survival in natural environments, degenerative disease, and aging. Understanding mechanisms of stress response dimorphism can therefore provide insights into sex-specific population dynamics, disease, and longevity.


Asunto(s)
Proteínas de Caenorhabditis elegans , Trastornos del Desarrollo Sexual , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Femenino , Longevidad , Masculino , Especies Reactivas de Oxígeno/metabolismo , Caracteres Sexuales
14.
Biochem Mol Biol Educ ; 48(5): 516-517, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32823376

RESUMEN

The rush to remote learning during the COVD-19 pandemic has caused instructors to rapidly adapt mechanisms of learning. Here, I describe an online concept mapping activity for membrane transport mechanisms that can be accomplished by students working together remotely and either synchronously or asynchronously.


Asunto(s)
Bioquímica/educación , COVID-19 , Membrana Celular , Educación a Distancia , Aprendizaje , Pandemias , Transporte Biológico Activo , Curriculum , Humanos , Estudiantes
15.
Mol Biol Evol ; 25(11): 2349-59, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18718920

RESUMEN

Cyclooxygenase (COX) produces prostaglandins in animals via the oxidation and reduction of arachidonic acid. Different types and numbers of COX genes have been found in corals, sea squirts, fishes, and tetrapods, but no study has used a comparative phylogenetic approach to investigate the evolutionary history of this complex gene family. Therefore, to examine COX evolution in the teleosts and chordates, 9 novel COX sequences (possessing residues and domains critical to COX function) were acquired from the euryhaline killifish, longhorn sculpin, sea lamprey, Atlantic hagfish, and amphioxus using standard polymerase chain reaction (PCR) and cloning methods. Phylogenetic analyses of these and other COX sequences show a complicated history of COX duplications and losses. There are three main lineages of COX in the chordates corresponding to the three subphyla in the phylum Chordata, with each lineage representing an independent COX duplication. Hagfish and lamprey most likely have traditional COX-1/2 genes, suggesting that these genes originated with the first round of genome duplication in the vertebrates according to the 2R hypothesis and are not exclusively present in the gnathostomes. All teleosts examined have three COX genes due to a teleost-specific genome duplication followed by variable loss of a COX-1 (in the zebrafish and rainbow trout) or COX-2 gene (in the derived teleosts). Future studies should examine the functional ramifications of these differential gene losses.


Asunto(s)
Cordados no Vertebrados/genética , Evolución Molecular , Peces/genética , Duplicación de Gen , Prostaglandina-Endoperóxido Sintasas/genética , Secuencias de Aminoácidos , Animales , Cordados no Vertebrados/clasificación , Cordados no Vertebrados/enzimología , Clonación Molecular , Femenino , Peces/clasificación , Fundulidae , Anguila Babosa , Humanos , Lampreas , Filogenia , Prostaglandina-Endoperóxido Sintasas/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Análisis de Secuencia de Proteína
16.
Genetics ; 208(4): 1467-1482, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29487136

RESUMEN

Extracellular matrix barriers and inducible cytoprotective genes form successive lines of defense against chemical and microbial environmental stressors. The barrier in nematodes is a collagenous extracellular matrix called the cuticle. In Caenorhabditis elegans, disruption of some cuticle collagen genes activates osmolyte and antimicrobial response genes. Physical damage to the epidermis also activates antimicrobial responses. Here, we assayed the effect of knocking down genes required for cuticle and epidermal integrity on diverse cellular stress responses. We found that disruption of specific bands of collagen, called annular furrows, coactivates detoxification, hyperosmotic, and antimicrobial response genes, but not other stress responses. Disruption of other cuticle structures and epidermal integrity does not have the same effect. Several transcription factors act downstream of furrow loss. SKN-1/Nrf and ELT-3/GATA are required for detoxification, SKN-1/Nrf is partially required for the osmolyte response, and STA-2/Stat and ELT-3/GATA for antimicrobial gene expression. Our results are consistent with a cuticle-associated damage sensor that coordinates detoxification, hyperosmotic, and antimicrobial responses through overlapping, but distinct, downstream signaling.


Asunto(s)
Caenorhabditis elegans/fisiología , Ambiente , Estrés Fisiológico , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Interacción Gen-Ambiente , Secuenciación de Nucleótidos de Alto Rendimiento , Inactivación Metabólica/genética , Ósmosis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , Transgenes
17.
FEBS J ; 274(22): 5782-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17944943

RESUMEN

Osmotic homeostasis is a fundamental requirement for life. In general, the effector mechanisms that mediate cellular and extracellular osmoregulation in animals are reasonably well defined. However, at the molecular level, little is known about how animals detect osmotic and ionic perturbations and transduce them into regulatory responses. The nematode Caenorhabditis elegans provides numerous powerful experimental advantages for defining the genes and integrated gene networks that underlie basic biological processes. These advantages include a fully sequenced and well-annotated genome, forward and reverse genetic and molecular tractability, and a relatively simple anatomy. C. elegans normally inhabits soil environments where it is exposed to repeated osmotic stress. In the laboratory, nematodes readily acclimate to and recover from extremes of hypertonicity. We review recent progress in defining the molecular mechanisms that underlie osmosensing and associated signal transduction in C. elegans. Some of these mechanisms are now known to be highly conserved. Therefore, studies of osmosensing in nematodes have provided, and will undoubtedly continue to provide, new insights into similar processes in more complex organisms including mammals.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Ósmosis , Transducción de Señal , Animales
18.
Genetics ; 206(2): 859-871, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28341649

RESUMEN

The transcription factor SKN-1 (Skinhead family member-1) in Caenorhabditis elegans is a homolog of the mammalian Nrf-2 protein and functions to promote oxidative stress resistance and longevity. SKN-1 mediates protection from reactive oxygen species (ROS) via the transcriptional activation of genes involved in antioxidant defense and phase II detoxification. Although many core regulators of SKN-1 have been identified, much remains unknown about this complex signaling pathway. We carried out an ethyl methanesulfonate (EMS) mutagenesis screen and isolated six independent mutants with attenuated SKN-1-dependent gene activation in response to acrylamide. All six were found to contain mutations in F46F11.6/xrep-4 (xenobiotics response pathways-4), which encodes an uncharacterized F-box protein. Loss of xrep-4 inhibits the skn-1-dependent expression of detoxification genes in response to prooxidants and decreases survival of oxidative stress, but does not shorten life span under standard culture conditions. XREP-4 interacts with the ubiquitin ligase component SKR-1 and the SKN-1 principal repressor WDR-23, and knockdown of xrep-4 increases nuclear localization of a WDR-23::GFP fusion protein. Furthermore, a missense mutation in the conserved XREP-4 F-box domain that reduces interaction with SKR-1 but not WDR-23 strongly attenuates SKN-1-dependent gene activation. These results are consistent with XREP-4 influencing the SKN-1 stress response by functioning as a bridge between WDR-23 and the ubiquitin ligase component SKR-1.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Proteínas F-Box/genética , Proteínas Nucleares/genética , Estrés Oxidativo/genética , Proteínas Ligasas SKP Cullina F-box/genética , Factores de Transcripción/genética , Animales , Caenorhabditis elegans/genética , Mutación Missense , Proteínas Represoras
19.
Artículo en Inglés | MEDLINE | ID: mdl-16814584

RESUMEN

Neuronal NOS (nNOS) is a constitutively expressed enzyme that catalyzes the oxidation of L-arginine and water to L-citrulline and the gas nitric oxide (NO). Nitric oxide is involved in regulation of a variety of processes, including: vascular tone, neurotransmission, and ion balance in mammals and fishes. In this study, we have cloned and characterized a putative NOS homologue from the brain of the euryhaline killifish, Fundulus heteroclitus. Killifish NOS has 75% amino acid identity to human nNOS, and phylogenetic analysis groups the killifish sequence with the mammalian nNOS, suggesting that it is a mammalian orthologue. Relative quantitative reverse transcriptase-PCR demonstrated that killifish nNOS mRNA is highly expressed in the brain and gill followed by the stomach, kidney, opercular epithelium, intestine and heart. Immunohistochemistry localized nNOS to nerve fibers and epithelial cells adjacent to mitochondrion-rich cells (ion transporting cell) in the gill, suggesting that nNOS production of NO may contribute to regulation of vascular tone and/or MRC function in the teleost gill.


Asunto(s)
Encéfalo/metabolismo , Fundulidae/metabolismo , Branquias/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Secuencia de Aminoácidos , Animales , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo I/genética , Especificidad de Órganos , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido
20.
Mech Ageing Dev ; 149: 88-98, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26056713

RESUMEN

The SKN-1/Nrf transcription factors are master regulators of oxidative stress responses and are emerging as important determinants of longevity. We previously identified a protein named WDR-23 as a direct repressor of SKN-1 in C. elegans. Loss of wdr-23 influences stress resistance, longevity, development, and reproduction, but it is unknown if WDR-23 influences development and reproduction solely through SKN-1 and the mechanisms by which SKN-1 promotes stress resistance and longevity are poorly defined. Here, we characterize phenotypes of wdr-23 and skn-1 manipulation and explore the role of glutathione. We provide evidence that diverse wdr-23 phenotypes are dependent on SKN-1, that beneficial and detrimental phenotypes of wdr-23 and skn-1 can be partially decoupled, and that SKN-1 activation delays degenerative tissue changes during aging. We also show that total glutathione levels are substantially elevated when the wdr-23/skn-1 pathway is activated and that skn-1 is required for preserving this cellular antioxidant during stress and aging. Alternatively, total glutathione was not elevated in worms with reduced insulin/IGF-1-like signaling or dietary restriction suggesting that SKN-1 ensures longevity via different mechanisms under these conditions. Lastly, genetic interaction data revise our understanding of which skn-1 variants are required for longevity during dietary restriction.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Glutatión/metabolismo , Longevidad , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Arsenitos/química , Tamaño Corporal , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Mutación , Fenotipo , Pigmentación , Interferencia de ARN , Proteínas Represoras/genética , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA