Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
PLoS Genet ; 18(9): e1010430, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36166480

RESUMEN

Partial loss-of-function mutations in glycosylation pathways underlie a set of rare diseases called Congenital Disorders of Glycosylation (CDGs). In particular, DPAGT1-CDG is caused by mutations in the gene encoding the first step in N-glycosylation, DPAGT1, and this disorder currently lacks effective therapies. To identify potential therapeutic targets for DPAGT1-CDG, we performed CRISPR knockout screens in Drosophila cells for genes associated with better survival and glycoprotein levels under DPAGT1 inhibition. We identified hundreds of candidate genes that may be of therapeutic benefit. Intriguingly, inhibition of the mannosyltransferase Dpm1, or its downstream glycosylation pathways, could rescue two in vivo models of DPAGT1 inhibition and ER stress, even though impairment of these pathways alone usually causes CDGs. While both in vivo models ostensibly cause cellular stress (through DPAGT1 inhibition or a misfolded protein), we found a novel difference in fructose metabolism that may indicate glycolysis as a modulator of DPAGT1-CDG. Our results provide new therapeutic targets for DPAGT1-CDG, include the unique finding of Dpm1-related pathways rescuing DPAGT1 inhibition, and reveal a novel interaction between fructose metabolism and ER stress.


Asunto(s)
Trastornos Congénitos de Glicosilación , Manosiltransferasas , N-Acetilglucosaminiltransferasas/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Trastornos Congénitos de Glicosilación/genética , Fructosa , Genoma , Glicoproteínas/genética , Humanos , Manosiltransferasas/genética
2.
Proc Natl Acad Sci U S A ; 116(44): 22322-22330, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31611372

RESUMEN

Early host responses toward pathogens are essential for defense against infection. In Caenorhabditis elegans, the transcription factor, SKN-1, regulates cellular defenses during xenobiotic intoxication and bacterial infection. However, constitutive activation of SKN-1 results in pleiotropic outcomes, including a redistribution of somatic lipids to the germline, which impairs health and shortens lifespan. Here, we show that exposing C. elegans to Pseudomonas aeruginosa similarly drives the rapid depletion of somatic, but not germline, lipid stores. Modulating the epigenetic landscape refines SKN-1 activity away from innate immunity targets, which alleviates negative metabolic outcomes. Similarly, exposure to oxidative stress redirects SKN-1 activity away from pathogen response genes while restoring somatic lipid distribution. In addition, activating p38/MAPK signaling in the absence of pathogens, is sufficient to drive SKN-1-dependent loss of somatic fat. These data define a SKN-1- and p38-dependent axis for coordinating pathogen responses, lipid homeostasis, and survival and identify transcriptional redirection, rather than inactivation, as a mechanism for counteracting the pleiotropic consequences of aberrant transcriptional activity.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Metabolismo de los Lípidos , Infecciones por Pseudomonas/genética , Factores de Transcripción/metabolismo , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Inmunidad Innata , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Factores de Transcripción/genética , Transcriptoma , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
PLoS Genet ; 14(7): e1007520, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30020921

RESUMEN

Across organisms, manipulation of biosynthetic capacity arrests development early in life, but can increase health- and lifespan post-developmentally. Here we demonstrate that this developmental arrest is not sickness but rather a regulated survival program responding to reduced cellular performance. We inhibited protein synthesis by reducing ribosome biogenesis (rps-11/RPS11 RNAi), translation initiation (ifg-1/EIF3G mutation and egl-45/EIF3A RNAi), or ribosome progression (cycloheximide treatment), all of which result in a specific arrest at larval stage 2 of C. elegans development. This quiescent state can last for weeks-beyond the normal C. elegans adult lifespan-and is reversible, as animals can resume reproduction and live a normal lifespan once released from the source of protein synthesis inhibition. The arrest state affords resistance to thermal, oxidative, and heavy metal stress exposure. In addition to cell-autonomous responses, reducing biosynthetic capacity only in the hypodermis was sufficient to drive organism-level developmental arrest and stress resistance phenotypes. Among the cell non-autonomous responses to protein synthesis inhibition is reduced pharyngeal pumping that is dependent upon AMPK-mediated signaling. The reduced pharyngeal pumping in response to protein synthesis inhibition is recapitulated by exposure to microbes that generate protein synthesis-inhibiting xenobiotics, which may mechanistically reduce ingestion of pathogen and toxin. These data define the existence of a transient arrest-survival state in response to protein synthesis inhibition and provide an evolutionary foundation for the conserved enhancement of healthy aging observed in post-developmental animals with reduced biosynthetic capacity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas Quinasas Activadas por AMP/genética , Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Animales , Evolución Biológica , Proteínas de Caenorhabditis elegans/genética , Conducta Alimentaria/fisiología , Larva/fisiología , Longevidad/efectos de los fármacos , Longevidad/genética , Faringe/fisiología , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Interferencia de ARN , Ribosomas/efectos de los fármacos , Ribosomas/genética , Ribosomas/metabolismo , Transducción de Señal/genética , Tejido Subcutáneo/metabolismo
4.
Proc Natl Acad Sci U S A ; 112(50): 15378-83, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621724

RESUMEN

Animals in nature are continually challenged by periods of feast and famine as resources inevitably fluctuate, and must allocate somatic reserves for reproduction to abate evolutionary pressures. We identify an age-dependent lipid homeostasis pathway in Caenorhabditis elegans that regulates the mobilization of lipids from the soma to the germline, which supports fecundity but at the cost of survival in nutrient-poor and oxidative stress environments. This trade-off is responsive to the levels of dietary carbohydrates and organismal oleic acid and is coupled to activation of the cytoprotective transcription factor SKN-1 in both laboratory-derived and natural isolates of C. elegans. The homeostatic balance of lipid stores between the somatic and germ cells is mediated by arachidonic acid (omega-6) and eicosapentaenoic acid (omega-3) precursors of eicosanoid signaling molecules. Our results describe a mechanism for resource reallocation within intact animals that influences reproductive fitness at the cost of somatic resilience.


Asunto(s)
Caenorhabditis elegans/metabolismo , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/farmacología , Alimentos , Células Germinativas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Animales , Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/efectos de los fármacos , Ácido Oléico/deficiencia , Reproducción/efectos de los fármacos , Análisis de Supervivencia , Vitelogénesis/efectos de los fármacos
5.
Mamm Genome ; 25(5-6): 202-10, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700286

RESUMEN

G protein-coupled receptors strongly modulate neuronal excitability but there has been little evidence for G protein mechanisms in genetic epilepsies. Recently, four patients with epileptic encephalopathy (EIEE17) were found to have mutations in GNAO1, the most abundant G protein in brain, but the mechanism of this effect is not known. The GNAO1 gene product, Gαo, negatively regulates neurotransmitter release. Here, we report a dominant murine model of Gnao1-related seizures and sudden death. We introduced a genomic gain-of-function knock-in mutation (Gnao1 (+/G184S)) that prevents Go turnoff by Regulators of G protein signaling proteins. This results in rare seizures, strain-dependent death between 15 and 40 weeks of age, and a markedly increased frequency of interictal epileptiform discharges. Mutants on a C57BL/6J background also have faster sensitization to pentylenetetrazol (PTZ) kindling. Both premature lethality and PTZ kindling effects are suppressed in the 129SvJ mouse strain. We have mapped a 129S-derived modifier locus on Chromosome 17 (within the region 41-70 MB) as a Modifer of G protein Seizures (Mogs1). Our mouse model suggests a novel gain-of-function mechanism for the newly defined subset of epileptic encephalopathy (EIEE17). Furthermore, it reveals a new epilepsy susceptibility modifier Mogs1 with implications for the complex genetics of human epilepsy as well as sudden death in epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mutación , Animales , Encéfalo/metabolismo , Encéfalo/patología , Epilepsia/mortalidad , Epilepsia/patología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Genetics ; 214(4): 913-925, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32047096

RESUMEN

Endoplasmic reticulum (ER) stress-induced apoptosis is a primary cause and modifier of degeneration in a number of genetic disorders. Understanding how genetic variation influences the ER stress response and subsequent activation of apoptosis could improve individualized therapies and predictions of outcomes for patients. In this study, we find that the uncharacterized, membrane-bound metallopeptidase CG14516 in Drosophila melanogaster, which we rename as SUPpressor of ER stress-induced DEATH (superdeath), plays a role in modifying ER stress-induced apoptosis. We demonstrate that loss of superdeath reduces apoptosis and degeneration in the Rh1G69D model of ER stress through the JNK signaling cascade. This effect on apoptosis occurs without altering the activation of the unfolded protein response (IRE1 and PERK), suggesting that the beneficial prosurvival effects of this response are intact. Furthermore, we show that superdeath functions epistatically upstream of CDK5-a known JNK-activated proapoptotic factor in this model of ER stress. We demonstrate that superdeath is not only a modifier of this particular model, but affects the general tolerance to ER stress, including ER stress-induced apoptosis. Finally, we present evidence of Superdeath localization to the ER membrane. While similar in sequence to a number of human metallopeptidases found in the plasma membrane and ER membrane, its localization suggests that superdeath is orthologous to ERAP1/2 in humans. Together, this study provides evidence that superdeath is a link between stress in the ER and activation of cytosolic apoptotic pathways.


Asunto(s)
Apoptosis , Proteínas de Drosophila/metabolismo , Estrés del Retículo Endoplásmico , N-Acetilgalactosaminiltransferasas/metabolismo , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Epistasis Genética , N-Acetilgalactosaminiltransferasas/genética , Respuesta de Proteína Desplegada
7.
J Gerontol A Biol Sci Med Sci ; 74(8): 1189-1197, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-30828708

RESUMEN

Air pollution is a heterogeneous environmental toxicant that impacts humans throughout their life. We introduce Caenorhabditis elegans as a valuable air pollution model with its short lifespan, medium-throughput capabilities, and highly conserved biological pathways that impact healthspan. We exposed developmental and adult life stages of C. elegans to airborne nano-sized particulate matter (nPM) produced by traffic emissions and measured biological and molecular endpoints that changed in response. Acute nPM did not cause lethality in C. elegans, but short-term exposure during larval stage 1 caused delayed development. Gene expression responses to nPM exposure overlapped with responses of mouse and cell culture models of nPM exposure in previous studies. We showed further that the skn-1/Nrf2 antioxidant response has a role in the development and hormetic effects of nPM. This study introduces the worm as a new resource and complementary model for mouse and cultured cell systems to study air pollution toxicity across the lifespan.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Longevidad/efectos de los fármacos , Material Particulado/toxicidad , Emisiones de Vehículos/toxicidad , Animales , Tamaño Corporal/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Modelos Animales , Nanopartículas , Faringe/efectos de los fármacos , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA