Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Neurobiol Dis ; 152: 105278, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33516872

RESUMEN

Machado-Joseph disease (MJD) or Spinocerebellar ataxia type 3 (SCA3) is a progressive neurodegenerative disorder that affects movement coordination leading to a premature death. Despite several efforts, no disease-modifying treatment is yet available for this disease. Previous studies pinpointed the modulation of serotonergic signaling, through pharmacological inhibition of the serotonin transporter SERT, as a promising therapeutic approach for MJD/SCA3. Here, we describe the 5-HT1A receptor as a novel therapeutic target in MJD, using a C. elegans model of ATXN3 proteotoxicity. Chronic and acute administration of befiradol (also known as NLX-112), a highly specific 5-HT1A agonist, rescued motor function and suppressed mutant ATXN3 aggregation. This action required the 5-HT1A receptor orthologue in the nematode, SER-4. Tandospirone, a clinically tested 5-HT1A receptor partial agonist, showed a limited impact on animals' motor dysfunction on acute administration and a broader receptor activation profile upon chronic treatment, its effect depending on 5-HT1A but also on the 5-HT6/SER-5 and 5-HT7/SER-7 receptors. Our results support high potency and specificity of befiradol for activation of 5-HT1A/SER-4 receptors and highlight the contribution of the auto- and hetero-receptor function to the therapeutic outcome in this MJD model. Our study deepens the understanding of serotonergic signaling modulation in the suppression of ATXN3 proteotoxicity and suggests that a potent and selective 5-HT1A receptor agonist such as befiradol could constitute a promising therapeutic agent for MJD.


Asunto(s)
Enfermedad de Machado-Joseph , Piperidinas/farmacología , Piridinas/farmacología , Receptor de Serotonina 5-HT1A/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Animales , Ataxina-3/efectos de los fármacos , Ataxina-3/genética , Ataxina-3/metabolismo , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Mutación , Agregación Patológica de Proteínas
2.
PLoS Pathog ; 15(2): e1007598, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30759156

RESUMEN

Resistance to the anthelmintic macrocyclic lactone ivermectin (IVM) has a great impact on the control of parasitic nematodes. The mechanisms by which nematodes adapt to IVM remain to be deciphered. We have identified NHR-8, a nuclear hormone receptor involved in the xenobiotic response in Caenorhabditis elegans, as a new regulator of tolerance to IVM. Loss-of-function nhr-8(ok186) C. elegans mutants subjected to larval development assays and electropharyngeogram measurements, displayed hypersensitivity to IVM, and silencing of nhr-8 in IVM-resistant worms increased IVM efficacy. In addition, compared to wild-type worms, nhr-8 mutants under IVM selection pressure failed to acquire tolerance to the drug. In addition, IVM-hypersensitive nhr-8(ok186) worms displayed low transcript levels of several genes from the xenobiotic detoxification network and a concomitant low Pgp-mediated drug efflux activity. Interestingly, some pgp and cyp genes known to impact IVM tolerance in many nematode species, were down regulated in nhr-8 mutants and inversely upregulated in IVM-resistant worms. Moreover, pgp-6 overexpression in nhr-8(ok186) C. elegans increased tolerance to IVM. Importantly, NHR-8 function was rescued in nhr-8(ok186) C. elegans with the homolog of the parasitic nematode Haemonchus contortus, and silencing of Hco-nhr-8 by RNAi on L2 H. contortus larvae increased IVM susceptibility in both susceptible and resistant H. contortus isolates. Thus, our data show that NHR-8 controls the tolerance and development of resistance to IVM in C. elegans and the molecular basis for this relates to the NHR-8-mediated upregulation of IVM detoxification genes. Since our results show that Hco-nhr-8 functions similarly to Cel-nhr-8, this study helps to better understand mechanisms underlying failure in drug efficacy and open perspectives in finding new compounds with NHR-8 antagonist activity to potentiate IVM efficacy.


Asunto(s)
Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Ivermectina/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Antihelmínticos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiología , Resistencia a Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Haemonchus , Ivermectina/farmacología , Larva , Infecciones por Nematodos/virología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/efectos de los fármacos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/efectos de los fármacos
3.
Molecules ; 26(6)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33802064

RESUMEN

Caffeic and dihydrocaffeic acid are relevant microbial catabolites, being described as products from the degradation of different phenolic compounds i.e., hydroxycinnamoyl derivatives, anthocyanins or flavonols. Furthermore, caffeic acid is found both in free and esterified forms in many fruits and in high concentrations in coffee. These phenolic acids may be responsible for a part of the bioactivity associated with the intake of phenolic compounds. With the aim of progressing in the knowledge of the health effects and mechanisms of action of dietary phenolics, the model nematode Caenorhabditis elegans has been used to evaluate the influence of caffeic and dihydrocaffeic acids on lifespan and the oxidative stress resistance. The involvement of different genes and transcription factors related to longevity and stress resistance in the response to these phenolic acids has also been explored. Caffeic acid (CA, 200 µM) and dihydrocaffeic acid (DHCA, 300 µM) induced an increase in the survival rate of C. elegans under thermal stress. Both compounds also increased the mean and maximum lifespan of the nematode, compared to untreated worms. In general, treatment with these acids led to a reduction in intracellular ROS concentrations, although not always significant. Results of gene expression studies conducted by RT-qPCR showed that the favorable effects of CA and DHCA on oxidative stress and longevity involve the activation of several genes related to insulin/IGF-1 pathway, such as daf-16, daf-18, hsf-1 and sod-3, as well as a sirtuin gene (sir-2.1).


Asunto(s)
Ácidos Cafeicos/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Antioxidantes/farmacología , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Ácidos Cafeicos/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Longevidad/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/genética , Factores de Transcripción/efectos de los fármacos
4.
J Neurosci ; 39(21): 4142-4152, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30886012

RESUMEN

Cannabis sativa alters sensory perception and exhibits potential medicinal benefits. In mammals, cannabinoids activate two canonical receptors, CB1/CB2, as well additional receptors/ion channels whose overall contributions to cannabinoid signaling have yet to be fully assessed. In Caenorhabditis elegans, the endogenous cannabinoid receptor agonist, 2-arachidonoylglycerol (2-AG) activates a CB1 ortholog, NPR-19, to modulate behavior (Oakes et al., 2017). In addition, 2-AG stimulates the NPR-19 independent release of both serotonin (5-HT) and dopamine (DA) from subsets of monoaminergic neurons to modulate locomotory behaviors through a complex monoaminergic signaling pathway involving multiple serotonin and dopamine receptors. 2-AG also inhibits locomotion in remodeled monoamine receptor mutant animals designed to measure the acute release of either 5-HT or DA, confirming the direct effects of 2-AG on monoamine release. 2-AG-dependent locomotory inhibition requires the expression of transient receptor potential vanilloid 1 (TRPV1) and TRPN-like channels in the serotonergic or dopaminergic neurons, respectively, and the acute pharmacological inhibition of the TRPV1-like channel abolishes both 2-AG-dependent 5-HT release and locomotory inhibition, suggesting the 2-AG may activate the channel directly. This study highlights the advantages of identifying and assessing both CB1/CB2-dependent and independent cannabinoid signaling pathways in a genetically tractable, mammalian predictive model, where cannabinoid signaling at the molecular/neuronal levels can be correlated directly with changes in behavior.SIGNIFICANCE STATEMENT This study is focused on assessing CB1/CB2-independent cannabinoid signaling in a genetically tractable, whole-animal model where cannabinoid signaling at the molecular/neuronal levels can be correlated with behavioral change. Caenorhabditis elegans contains a cannabinoid signaling system mediated by a canonical cannabinoid receptor, NPR-19, with orthology to human CB1/CB2 (Oakes et al., 2017). The present study has characterized an NPR-19-independent signaling pathway that involves the cannabinoid-dependent release of both serotonin and dopamine and the expression of distinct TRP-like channels on the monoaminergic neurons. Our work should be of interest to those studying the complexities of CB1/CB2-independent cannabinoid signaling, the role of TRP channels in the modulation of monoaminergic signaling, and the cannabinoid-dependent modulation of behavior.


Asunto(s)
Cannabinoides/farmacología , Dopamina/metabolismo , Serotonina/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Ácidos Araquidónicos/farmacología , Conducta Animal , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Agonistas de Receptores de Cannabinoides/farmacología , Endocannabinoides/farmacología , Glicéridos/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Canales Catiónicos TRPV/efectos de los fármacos
5.
Eur J Nutr ; 59(1): 137-150, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30710163

RESUMEN

PURPOSE: Hibiscus sabdariffa L. is commonly used as an ingredient for herbal teas and food supplements. Several studies demonstrated the beneficial effects of Hibiscus sabdariffa L. extracts (HSE); however, the bioactive components and their mode of action still remain unclear. Caenorhabditis elegans (C. elegans) was used to study health-related effects and the underlying molecular mechanisms of HSE in this model organism as well as effects of hydroxycitric acid (HCA), a main compound of HSE, and its structural analogue isocitric acid (ICA). METHODS: Survival and locomotion were detected by touch-provoked movement. Thermotolerance was analysed using the nucleic acid stain SYTOX green, and intracellular ROS accumulation was measured via oxidation of H2DCF. Localisation of the transcription factors DAF-16 and SKN-1 was analysed in transgenic strains (DAF-16::GFP, SKN-1::GFP). The involvement of DAF-16 and SKN-1 was further investigated using loss-of-function strains as well as gene silencing by feeding RNAi-inducing bacteria. Protection against amyloid-ß toxicity was analysed using a transgenic strain with an inducible expression of human amyloid-ß peptides in body wall muscle cells (paralysis assay). RESULTS: HSE treatment resulted in a prominent extension of lifespan (up to 24%) and a reduction of the age-dependent decline in locomotion. HCA, a main compound of HSE increased lifespan too, but to a lesser extent (6%) while ICA was not effective. HSE and HCA did not modulate resistance against thermal stress conditions and did not exert antioxidative effects: HSE rather increased intracellular ROS levels, suggesting a pro-oxidative effect of the extract in vivo. HSE and HCA increased the nuclear localisation of the pivotal transcription factors DAF-16 and SKN-1 indicating an activation of these factors. Consistent with this result, lifespan prolongation by HSE was dependent on both transcription factors. In addition to the positive effect on lifespan, HSE treatment also elicited a (strong) protection against amyloid-ß induced toxicity in C. elegans in a DAF-16- and SKN-1-dependent manner. CONCLUSION: Our results demonstrate that HSE increases lifespan and protects against amyloid-ß toxicity in the model organism C. elegans. These effects were mediated, at least in parts via modulation of pathways leading to activation/nuclear localisation of DAF-16 and SKN-1. Since HCA, a main component of HSE causes only minor effects, additional bioactive compounds like flavonoids or anthocyanins as well as synergistic effects of these compounds should be investigated.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/metabolismo , Hibiscus , Longevidad/fisiología , Extractos Vegetales/farmacología , Factores de Transcripción/metabolismo , Animales , Antioxidantes/farmacología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/genética , Longevidad/efectos de los fármacos , Modelos Animales , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
6.
Toxicol Appl Pharmacol ; 376: 1-8, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31100289

RESUMEN

Unusual cases of fatal lung injury, later determined to be a result of exposure to chemicals used as humidifier disinfectants, were reported among Korean children from 2006 to 2011. This resulted in considerable study of the pulmonary toxicity of humidifier disinfectant chemicals to establish the causal relationship between exposure and lung disease. However, the systemic toxicity of the former and health effects other than lung disease are not fully understood. Here, we investigated the effect of 5-chloro-2-methyl-4-isothiazoline-3-one and 2-methyl-4-isothiazolin-3-one (CMIT/MIT), among the humidifier disinfectants used in the accidents, on the development of metabolic toxicity in the model organism, Caenorhabditis elegans using an exposure scenario comparison. We screened the potential of CMIT/MIT to induce metabolic toxicity using C. elegans oga-1(ok1207) and ogt-1(ok1474) mutants. We also performed a pathway analysis based on C. elegans transcription factor RNAi library screening to identify the underlying toxicity mechanisms. Finally, to understand the critical window of exposure for metabolic toxicity, responses to exposure during different periods in the life cycles of the worms were compared. We determined that CMIT/MIT could induce metabolic toxicity through O-linked N-acetylglucosamine transferase and early life seems to be the critical window for exposure for metabolic toxicity for this substance. The O-linked N-acetylglucosamine transferase pathway is conserved from worms to humans; our results thus insinuate that early-life exposure to CMIT/MIT could cause metabolic health problems during adult life in humans. We therefore suggest that a systemic toxicity approach should be considered to comprehensively understand the adverse health effects of humidifier disinfectant misuse.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Desinfectantes/toxicidad , N-Acetilglucosaminiltransferasas/fisiología , Tiazoles/toxicidad , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Humidificadores , Mutación , N-Acetilglucosaminiltransferasas/genética , Estrés Oxidativo/efectos de los fármacos , Interferencia de ARN/efectos de los fármacos
7.
Gerontology ; 65(5): 513-523, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31112974

RESUMEN

BACKGROUND: With the acceleration of aging process in human society, improvements of the physical functionality and life quality in the elderly population are more meaningful than pure longevity. Buckwheat trypsin inhibitor is a low molecular weight polypeptide extracted from buckwheat, which is a beneficial food for improving the health in the elderly. OBJECTIVES: The aim of the current study was to evaluate the potential beneficial effects of recombinant buckwheat trypsin inhibitor (rBTI) on age-dependent function decline and the primary mechanism. METHOD: Day 10 N2 Caenorhabditis elegans and day 6 AM140 C. elegans cultured at 25°C were used as models of aging and age-related disease, respectively. Motor function was as an indicator of age-dependent function. ATP content and damage mitochondrial DNA mass were detected to assess mitochondrial damage and function by ATP Assay Kit and agarose gel electrophoresis, respectively. Soluble protein content was quantified by SDS polyacrylamide gel electrophoresis. Autophagy-related genes transcription levels, autophagy marker proteins lgg-1, and lysosomal content were analyzed to quantify autophagy levels by qRT-PCR, transgenic C. elegans, and lysosomal staining. Autophagy inhibitor chloroquine, daf-16 mutant, and RNA Interference were used to determine the roles of autophagy and DAF-16 in rBTI-mediated effects. RESULTS: In this study, we found that rBTI could decrease the proportions of insoluble protein and impaired mitochondria, finally reduce motility deficits in both models. Further study indicated that rBTI activated the autophagy, and the inhibition of autophagy reduced rBTI-mediated beneficial effects. Genetic analyses showed the transcriptional activity of DAF-16 was increased by rBTI and was required for rBTI-mediated beneficial effects. CONCLUSIONS: These data indicated that rBTI might promote the autophagy to alleviate the age-related functional decline via DAF-16 in C. elegans and suggested a potential role of rBTI as a nutraceutical for the improvement of age-related complications.


Asunto(s)
Envejecimiento/efectos de los fármacos , Autofagia/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteínas de Plantas/farmacología , Proteostasis/efectos de los fármacos , Inhibidores de Tripsina/farmacología , Adenosina Trifosfato/metabolismo , Amebicidas/farmacología , Animales , Autofagia/genética , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cloroquina/farmacología , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/metabolismo , Modelos Animales de Enfermedad , Fagopyrum , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Homeostasis/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Recombinantes
8.
Int J Food Sci Nutr ; 70(5): 595-602, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30624146

RESUMEN

The present investigation demonstrates the longevity-promoting effects of 3-methyl-3-buten-1-ol (isoprenol) in the animal model Caenorhabditis elegans that might be served as a lead nutraceutical in geriatric research. Our results showed that 0.5 mM isoprenol extended the mean lifespan of worms by 25% in comparison to control worms. Isoprenol also significantly enhanced survival of the worms under various stress conditions. It was found that the longevity-promoting effects of isoprenol were associated with improved age-associated physiological behaviour and reduced intracellular reactive oxygen species (ROS) accumulation. Finally, studies with gene-specific mutants revealed the involvement of pro-longevity transcription factors (TFs) DAF-16 and SKN-1 with simultaneous over-expression of GST-4 and SOD-3 in isoprenol treated worms. In silico analysis revealed the binding affinity of isoprenol with DAF-16 and SKN-1 TFs. Together, the findings suggest that isoprenol is able to enhance the lifespan of C. elegans and embarks its potential in the developments of formulations for age-related ailments.


Asunto(s)
Butanoles/farmacología , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/crecimiento & desarrollo , Longevidad/efectos de los fármacos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Modelos Animales , Simulación del Acoplamiento Molecular , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
9.
Molecules ; 24(5)2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30845642

RESUMEN

Endopleura uchi (Huber) Cuatrec (Humiriaceae), known as uxi or uxi-amarelo in Brazil, is an endemic tree of the Amazon forest. In traditional medicine, its stem bark is used to treat a variety of health disorders, including cancer, diabetes, arthritis, uterine inflammation, and gynecological infections. According to HPLC analysis, the main constituent of the bark extract is the polyphenol bergenin. In the current study, we demonstrate by in vitro and in vivo experiments the antioxidant potential of a water extract from the stem bark of E. uchi. When tested in the model organism Caenorhabditis elegans, the extract enhanced stress resistance via the DAF-16/FOXO pathway. Additionally, the extract promoted an increase in the lifespan of the worms independent from caloric restriction. It also attenuated the age-related muscle function decline and formation of polyQ40 plaques, as a model for Huntington's disease. Thus, these data support anti-aging and anti-oxidant properties of E. uchi, which has not yet been described. More studies are needed to assess the real benefits of E. uchi bark for human health and its toxicological profile.


Asunto(s)
Benzopiranos/farmacología , Caenorhabditis elegans/efectos de los fármacos , Longevidad/efectos de los fármacos , Phaeophyceae/química , Corteza de la Planta/química , Animales , Antiinfecciosos/química , Antiinfecciosos/farmacología , Antioxidantes/farmacología , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Cromatografía Líquida de Alta Presión/métodos , Regulación de la Expresión Génica , Humanos , Enfermedad de Huntington/metabolismo , Proteínas de Insectos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenol/química , Fenol/farmacología , Extractos Vegetales/farmacología , Transducción de Señal
10.
Biochemistry ; 57(5): 722-731, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29319298

RESUMEN

Calumenin (Calu) is a well-conserved multi-EF-hand-containing Ca2+-binding protein. In this work, we focused on the alterations that calumenin has undergone during evolution. We demonstrate that vertebrate calumenin is significantly different from its invertebrate homologues with respect to its response to Ca2+ binding. Human calumenin (HsCalu1) is intrinsically unstructured in the Ca2+ free form and responds to Ca2+ with a dramatic gain in structure. Calumenin from Caenorhabditis elegans (CeCalu) is structured even in the apo form, with no conformational change upon binding of Ca2+. We decode this structural and functional distinction by identifying a single "Leu" residue-based switch located in the fourth EF-hand of HsCalu1, occupied by "Gly" in the invertebrate homologues. We demonstrate that replacing Leu with Gly (L150G) in HsCalu1 enables the protein to adopt a structural fold even in the Ca2+ free form, similar to CeCalu, leading to ligand compensation (adoption of structure in the absence of Ca2+). The fourth (of seven) EF-hand of HsCalu1 nucleates the structural fold of the protein depending on the switch residue (Gly or Leu). Our analyses reveal that the Leu that replaced Gly from fishes onward is absolutely conserved in higher vertebrates, while lower organisms have Gly, not only enlarging the scope of Ca2+-dependent structural transitions but also drawing a boundary between the invertebrate and vertebrate calumenin. The evolutionary selection of the switch residue strongly corroborates the change in the structure of the protein and its pleiotropic functions and seems like it can be extended to the presence or absence of a heart in that organism.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Proteínas de Unión al Calcio/química , Calcio/farmacología , Glicina/química , Leucina/química , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Calcio/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/genética , Secuencia Conservada , Evolución Molecular , Humanos , Invertebrados/metabolismo , Modelos Moleculares , Filogenia , Unión Proteica , Dominios Proteicos , Pliegue de Proteína , Estructura Terciaria de Proteína/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Vertebrados/metabolismo
11.
EMBO Rep ; 15(6): 705-13, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24764321

RESUMEN

The mechanisms that coordinate the regulation of autophagy with developmental signaling during multicellular organism development remain largely unknown. Here, we show that impaired function of ribosomal protein RPL-43 causes an accumulation of SQST-1 aggregates in the larval intestine, which are removed upon autophagy induction. Using this model to screen for autophagy regulators, we identify 139 genes that promote autophagy activity upon inactivation. Various signaling pathways, including Sma/Mab TGF-ß signaling, lin-35/Rb signaling, the XBP-1-mediated ER stress response, and the ATFS-1-mediated mitochondrial stress response, regulate the expression of autophagy genes independently of the TFEB homolog HLH-30. Our study thus provides a framework for understanding the role of signaling pathways in regulating autophagy under physiological conditions.


Asunto(s)
Autofagia/fisiología , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Regulación de la Expresión Génica/genética , Morfogénesis/fisiología , Proteínas Ribosómicas/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Técnica del Anticuerpo Fluorescente , Mucosa Intestinal/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Proteínas Ribosómicas/metabolismo
12.
Brain ; 138(Pt 11): 3221-37, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26373603

RESUMEN

Polyglutamine diseases are a class of dominantly inherited neurodegenerative disorders for which there is no effective treatment. Here we provide evidence that activation of serotonergic signalling is beneficial in animal models of Machado-Joseph disease. We identified citalopram, a selective serotonin reuptake inhibitor, in a small molecule screen of FDA-approved drugs that rescued neuronal dysfunction and reduced aggregation using a Caenorhabditis elegans model of mutant ataxin 3-induced neurotoxicity. MOD-5, the C. elegans orthologue of the serotonin transporter and cellular target of citalopram, and the serotonin receptors SER-1 and SER-4 were strong genetic modifiers of ataxin 3 neurotoxicity and necessary for therapeutic efficacy. Moreover, chronic treatment of CMVMJD135 mice with citalopram significantly reduced ataxin 3 neuronal inclusions and astrogliosis, rescued diminished body weight and strikingly ameliorated motor symptoms. These results suggest that small molecule modulation of serotonergic signalling represents a promising therapeutic target for Machado-Joseph disease.


Asunto(s)
Ataxina-3/efectos de los fármacos , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Citalopram/farmacología , Gliosis/metabolismo , Cuerpos de Inclusión/efectos de los fármacos , Locomoción/efectos de los fármacos , Enfermedad de Machado-Joseph/metabolismo , Neuronas/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/metabolismo , Animales , Ataxina-3/metabolismo , Conducta Animal/efectos de los fármacos , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Transmisión Sináptica/efectos de los fármacos
13.
Korean J Anesthesiol ; 77(3): 384-391, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38356139

RESUMEN

BACKGROUND: The anti-oxidative, anti-inflammatory, and anti-apoptotic effects of erythropoietin may provide neuroprotective effects. Erythropoietin also modulates autophagy signaling that may play a role in anesthesia-induced neurotoxicity (AIN). Herein, we investigated whether AIN can be attenuated by the neuroprotective effect of erythropoietin in the Caenorhabditis elegans (C. elegans). METHODS: Synchronized worms were divided into the control, Iso, EPO, and EPO-Iso groups. The chemotaxis index (CI) was evaluated when they reached the young adult stage. The lgg-1::GFP-positive puncta per seam cell were used to determine the autophagic events. The erythropoietin-mediated pathway of autophagy was determined by measuring the genetic expression level of let-363, bec-1, atg-7, atg-5, and lgg-3. RESULTS: Increased lgg-1::GFP puncta were observed in the Iso, EPO, and EPO-Iso groups. In the Iso group, only the let-363 level decreased significantly as compared to that in the control group (P = 0.009). bec-1 (P < 0.001), atg-5 (P = 0.012), and lgg-3 (P < 0.001) were expressed significantly more in the EPO-Iso group than in the Iso groups. Repeated isoflurane exposure during development decreased the CI. Erythropoietin could restore the decreased CI by isoflurane significantly in the EPO-Iso group. CONCLUSIONS: Erythropoietin showed neuroprotective effects against AIN and modulated the autophagic pathway in C. elegans. This experimental evidence of erythropoietin-related neuroprotection against AIN may be correlated with the induced autophagic degradation process that was sufficient for handling enhanced autophagy induction in erythropoietin-treated worms.


Asunto(s)
Autofagia , Caenorhabditis elegans , Eritropoyetina , Fármacos Neuroprotectores , Animales , Caenorhabditis elegans/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/fisiología , Eritropoyetina/farmacología , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/prevención & control , Anestesia/métodos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/efectos de los fármacos
14.
J Neurosci ; 32(49): 17909-20, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23223309

RESUMEN

Activity-dependent changes in presynaptic function represent a critical mechanism by which synaptic strength is controlled. However, how changes in synaptic activity couple to presynaptic components to control synaptic vesicle release and recycling are poorly understood. Sphingosine kinase (SphK) is a sphingolipid metabolic enzyme whose activity-dependent recruitment to membrane regions within presynaptic terminals promotes neurotransmitter release. Here, we show that synaptic recruitment of SPHK-1, the SphK ortholog in Caenorhabditis elegans, is mediated by presynaptic calcium influx. Quantitative fluorescence imaging of live presynaptic terminals reveals that blocking presynaptic calcium influx reduces synaptic SPHK-1 abundance whereas increasing calcium influx increases SPHK-1 synaptic abundance. CALM-1, the calcium and integrin binding protein ortholog, colocalizes with SPHK-1 at release sites and regulates muscarinic-mediated synaptic SPHK-1 recruitment. We identify two additional sphingolipid metabolic enzymes that are concentrated at presynaptic terminals, and mutants lacking one of these, HYL-1/ceramide synthase, have defects in synaptic transmission and in synaptic vesicle cycling. Finally, we show that SPHK-1 activity is required for the recruitment of the priming protein UNC-13/Munc13 to presynaptic terminals following activation by muscarinic signaling. These findings suggest that calcium-dependent regulation of local S1P metabolism at synapses may be an important mechanism by which synaptic vesicle priming factors are recruited to release sites to promote synaptic transmission.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Terminales Presinápticos/metabolismo , Esfingolípidos/metabolismo , Aldicarb/farmacología , Animales , Arecolina/farmacología , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas Portadoras , Agonistas Colinérgicos/farmacología , Inhibidores de la Colinesterasa/farmacología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Mutación , Imagen Óptica/métodos , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo
15.
Aging Cell ; 21(1): e13518, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35032420

RESUMEN

Diet regulates complex life-history traits such as longevity. For optimal lifespan, organisms employ intricate adaptive mechanisms whose molecular underpinnings are less known. We show that Caenorhabditis elegans FLR-4 kinase prevents lifespan differentials on the bacterial diet having higher Vitamin B12 levels. The flr-4 mutants are more responsive to the higher B12 levels of Escherichia coli HT115 diet, and consequently, have enhanced flux through the one-carbon cycle. Mechanistically, a higher level of B12 transcriptionally downregulates the phosphoethanolamine methyltransferase pmt-2 gene, which modulates phosphatidylcholine (PC) levels. Pmt-2 downregulation activates cytoprotective gene expression through the p38-MAPK pathway, leading to increased lifespan only in the mutant. Evidently, preventing bacterial B12 uptake or inhibiting one-carbon metabolism reverses all the above phenotypes. Conversely, supplementation of B12 to E. coli OP50 or genetically reducing PC levels in the OP50-fed mutant extends lifespan. Together, we reveal how worms maintain adaptive capacity to diets having varying micronutrient content to ensure a normal lifespan.


Asunto(s)
Proteínas de Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/efectos de los fármacos , Dieta , Longevidad/efectos de los fármacos , Vitamina B 12/uso terapéutico , Animales , Vitamina B 12/farmacología
16.
Cell Metab ; 3(4): 237-45, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16581001

RESUMEN

Starvation activates MAPK in the pharyngeal muscles of C. elegans through a muscarinic acetylcholine receptor, Gqalpha, and nPKC as shown by the following results: (1) Starvation causes phosphorylation of MAPK in pharyngeal muscle. (2) In a sensitized genetic background in which Gqalpha signaling cannot be downregulated, activation of the pathway by a muscarinic agonist causes lethal changes in pharyngeal muscle function. Starvation has identical effects. (3) A muscarinic antagonist blocks the effects of starvation on sensitized muscle. (4) Mutations and drugs that block any step of signaling from the muscarinic receptor to MAPK also block the effects of starvation on sensitized muscle. (5) Overexpression of MAPK in wild-type pharyngeal muscle mimics the effects of muscarinic agonist and of starvation on sensitized muscle. We suggest that, during starvation, the muscarinic pathway to MAPK is activated to change the pharyngeal muscle physiology to enhance ingestion of food when food becomes available.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/fisiología , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Muscarínicos/fisiología , Acetilcolina/fisiología , Animales , Arecolina/farmacología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Agonistas Colinérgicos/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Conducta Alimentaria , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/fisiología , Regulación Enzimológica de la Expresión Génica , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Mutación , Faringe/efectos de los fármacos , Faringe/enzimología , Fenotipo , Proteína Quinasa C/fisiología , Receptores Muscarínicos/efectos de los fármacos , Inanición
17.
Dev Cell ; 10(4): 473-82, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16563875

RESUMEN

C. elegans diapause, gonadal outgrowth, and life span are regulated by a lipophilic hormone, which serves as a ligand to the nuclear hormone receptor DAF-12. A key step in hormone production is catalyzed by the CYP450 DAF-9, but the extent of the biosynthetic pathway is unknown. Here, we identify a conserved Rieske-like oxygenase, DAF-36, as a component in hormone metabolism. Mutants display larval developmental and adult aging phenotypes, as well as patterns of epistasis similar to that of daf-9. Larval phenotypes are potently reversed by crude lipid extracts, 7-dehydrocholesterol, and a recently identified DAF-12 sterol ligand, suggesting that DAF-36 works early in the hormone biosynthetic pathway. DAF-36 is expressed primarily within the intestine, a major organ of metabolic and endocrine control, distinct from DAF-9. These results imply that C. elegans hormone production has multiple steps and is distributed, and that it may provide one way that tissues register their current physiological state during organismal commitments.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/fisiología , Deshidrocolesteroles/farmacología , Longevidad/efectos de los fármacos , Oxigenasas/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Catálisis , Deshidrocolesteroles/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Larva/enzimología , Larva/fisiología , Ligandos , Longevidad/fisiología , Datos de Secuencia Molecular , Estructura Molecular , Mutación , Oxigenasas/química , Oxigenasas/genética , Fenotipo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Neurochem Int ; 147: 105047, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33872680

RESUMEN

Mammalian nuclear distribution genes encode proteins with essential roles in neuronal migration and brain formation during embryogenesis. The implication of human nuclear distribution genes, namely nudC and NDE1 (Nuclear Distribution Element 1)/NDEL1 (Nuclear Distribution Element-Like 1), in psychiatric disorders including schizophrenia and bipolar disorder, has been recently described. The partial loss of NDEL1 expression results in neuronal migration defects, while ndel1 null knockout (KO) leads to early embryonic lethality in mice. On the other hand, loss-of-function of the orthologs of nuclear distribution element genes (nud) in Caenorhabditis elegans renders viable worms and influences behavioral endophenotypes associated with dopaminergic and serotoninergic pathways. In the present work, we evaluated the role of nud genes in monoamine levels at baseline and after the treatment with typical or atypical antipsychotics. Dopamine, serotonin and octopamine levels were significantly lower in homozygous loss-of-function mutant worms KO for nud genes compared with wild-type (WT) C. elegans at baseline. While treatment with antipsychotics determined significant differences in monoamine levels in WT, the nud KO mutant worms appear to respond differently to the treatment. According to the best of our knowledge, we are the first to report the influence of nud genes in the monoamine levels changes in response to antipsychotic drugs, ultimately placing the nuclear distribution genes family at the cornerstone of pathways involved in the modulation of monoamines in response to different classes of antipsychotic drugs.


Asunto(s)
Antipsicóticos/farmacología , Monoaminas Biogénicas/metabolismo , Encéfalo/efectos de los fármacos , Proteínas Portadoras/efectos de los fármacos , Mutación/efectos de los fármacos , Animales , Encéfalo/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo
19.
Cell Metab ; 1(5): 343-54, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-16054081

RESUMEN

The transient receptor potential (TRP) channels are implicated in various cellular processes, including sensory signal transduction and electrolyte homeostasis. We show here that the GTL-1 and GON-2 TRPM channels regulate electrolyte homeostasis in the C. elegans intestine. GON-2 is responsible for a large outwardly rectifying current of intestinal cells, and its activity is tightly regulated by intracellular Mg(2+) levels, while GTL-1 mainly contributes to appropriate Mg(2+) responsiveness of the outwardly rectifying current. We also used nickel cytotoxicity to study the function of these channels. Both GON-2 and GTL-1 are necessary for intestinal uptake of nickel, but GTL-1 is continuously active while GON-2 is inactivated at higher Mg(2+) levels. This type of differential regulation of intestinal electrolyte absorption ensures a constant supply of electrolytes through GTL-1, while occasional bursts of GON-2 activity allow rapid return to normal electrolyte concentrations following physiological perturbations.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Electrólitos/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Canales Iónicos/fisiología , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Calcio/metabolismo , Intestinos/citología , Canales Iónicos/efectos de los fármacos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Magnesio/metabolismo , Mutación , Níquel/toxicidad , Filogenia , Transducción de Señal/fisiología
20.
J Biol Chem ; 284(39): 26493-501, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19635802

RESUMEN

Galectins are a family of beta-galactoside-binding proteins that are widely found among animal species and that regulate diverse biological phenomena. To study the biological function of glycolipid-binding galectins, we purified recombinant Caenorhabditis elegans galectins (LEC-1-11) and studied their binding to C. elegans glycolipids. We found that LEC-8 binds to glycolipids in C. elegans through carbohydrate recognition. It has been reported that Cry5B-producing Bacillus thuringiensis strains can infect C. elegans and that the C. elegans Cry5B receptor molecules are glycolipids. We found that Cry5B and LEC-8 bound to C. elegans glycolipid-coated plates in a dose-dependent manner and that Cry5B binding to glycolipids was inhibited by the addition of LEC-8. LEC-8 is usually expressed strongly in the pharyngeal-intestinal valve and intestinal-rectal valve and is expressed weakly in intestine. However, when C. elegans were fed Escherichia coli expressing Cry5B, intestinal LEC-8::EGFP protein levels increased markedly. In contrast, LEC-8::EGFP expression triggered by Cry5B was reduced in toxin-resistant C. elegans mutants, which had mutations in genes involved in biosynthesis of glycolipids. Moreover, the LEC-8-deficient mutant was more susceptible to Cry5B than wild-type worms. These results suggest that the glycolipid-binding lectin LEC-8 contributes to host defense against bacterial infection by competitive binding to target glycolipid molecules.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Galectinas/metabolismo , Glucolípidos/metabolismo , Animales , Bacillus thuringiensis/metabolismo , Bacillus thuringiensis/fisiología , Toxinas de Bacillus thuringiensis , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Cromatografía Líquida de Alta Presión , Endotoxinas/metabolismo , Endotoxinas/toxicidad , Ensayo de Inmunoadsorción Enzimática , Galectinas/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidad , Interacciones Huésped-Patógeno , Mucosa Intestinal/metabolismo , Mutación , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda