Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Adv ; 5(12): eaax0292, 2019 12.
Article in English | MEDLINE | ID: mdl-31840061

ABSTRACT

The mechanistic basis for the biogenesis of peptide hormones and growth factors is poorly understood. Here, we show that the conserved endoplasmic reticulum membrane translocon-associated protein α (TRAPα), also known as signal sequence receptor 1, plays a critical role in the biosynthesis of insulin. Genetic analysis in the nematode Caenorhabditis elegans and biochemical studies in pancreatic ß cells reveal that TRAPα deletion impairs preproinsulin translocation while unexpectedly disrupting distal steps in insulin biogenesis including proinsulin processing and secretion. The association of common intronic single-nucleotide variants in the human TRAPα gene with susceptibility to type 2 diabetes and pancreatic ß cell dysfunction suggests that impairment of preproinsulin translocation and proinsulin trafficking may contribute to the pathogenesis of type 2 diabetes.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Calcium-Binding Proteins/metabolism , Insulin/biosynthesis , Membrane Glycoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Peptide/metabolism , Animals , Endoplasmic Reticulum Stress , Insulin/metabolism , Insulin Secretion , Protein Precursors/metabolism , Recombinant Fusion Proteins/metabolism
2.
G3 (Bethesda) ; 8(11): 3421-3431, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30194090

ABSTRACT

A feature common to late onset proteinopathic disorders is an accumulation of toxic protein conformers and aggregates in affected tissues. In the search for potential drug targets, many studies used high-throughput screens to find genes that modify the cytotoxicity of misfolded proteins. A complement to this approach is to focus on strategies that use protein aggregation as a phenotypic readout to identify pathways that control aggregate formation and maintenance. Here we use natural variation between strains of budding yeast to genetically map loci that influence the aggregation of a polyglutamine-containing protein derived from a mutant form of huntingtin, the causative agent in Huntington disease. Linkage analysis of progeny derived from a cross between wild and laboratory yeast strains revealed two polymorphic loci that modify polyglutamine aggregation. One locus contains the gene RFU1 which modifies ubiquitination states of misfolded proteins targeted by the E3-ubiquitin ligase complex Rsp5 Activity of the Rsp5 complex, and the mammalian homolog NEDD4, are critical in maintaining protein homeostasis in response to proteomic stress. Our analysis also showed linkage of the aggregation phenotype to a distinct locus containing a gene encoding the Rsp5-interacting Bul2 protein. Allele-swap experiments validated the impact of both RFU1 and BUL2 on huntingtin aggregation. Furthermore, we found that the nematode Caenorhabditis elegans' ortholog of Rsp5, wwp-1, also negatively regulates polyglutamine aggregation. Knockdown of the NEDD4 in human cells likewise altered polyglutamine aggregation. Taken together, these results implicate conserved processes involving the ubiquitin regulation network that modify protein aggregation and provide novel therapeutic targets for polyglutamine and other protein folding diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Caenorhabditis elegans Proteins/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Peptides/physiology , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Caenorhabditis elegans , Genetic Variation , HEK293 Cells , Humans , Mutation , Saccharomycetales/physiology
3.
Nat Commun ; 8: 14256, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220799

ABSTRACT

Limiting the debilitating consequences of ageing is a major medical challenge of our time. Robust pharmacological interventions that promote healthy ageing across diverse genetic backgrounds may engage conserved longevity pathways. Here we report results from the Caenorhabditis Intervention Testing Program in assessing longevity variation across 22 Caenorhabditis strains spanning 3 species, using multiple replicates collected across three independent laboratories. Reproducibility between test sites is high, whereas individual trial reproducibility is relatively low. Of ten pro-longevity chemicals tested, six significantly extend lifespan in at least one strain. Three reported dietary restriction mimetics are mainly effective across C. elegans strains, indicating species and strain-specific responses. In contrast, the amyloid dye ThioflavinT is both potent and robust across the strains. Our results highlight promising pharmacological leads and demonstrate the importance of assessing lifespans of discrete cohorts across repeat studies to capture biological variation in the search for reproducible ageing interventions.


Subject(s)
Caenorhabditis/drug effects , Genetic Background , Longevity/drug effects , Organic Chemicals/pharmacology , Animals , Benzothiazoles , Caenorhabditis/classification , Caenorhabditis/genetics , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Dose-Response Relationship, Drug , Fertility/drug effects , Fertility/genetics , Longevity/genetics , Reproducibility of Results , Species Specificity , Thiazoles/pharmacology
4.
Development ; 144(7): 1273-1282, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28209779

ABSTRACT

Animals change developmental fates in response to external cues. In the nematode Caenorhabditis elegans, unfavorable environmental conditions induce a state of diapause known as dauer by inhibiting the conserved DAF-2 insulin-like signaling (ILS) pathway through incompletely understood mechanisms. We have previously established a role for the C. elegans dosage compensation protein DPY-21 in the control of dauer arrest and DAF-2 ILS. Here, we show that the histone H4 lysine 20 methyltransferase SET-4, which also influences dosage compensation, promotes dauer arrest in part by repressing the X-linked ins-9 gene, which encodes a new agonist insulin-like peptide (ILP) expressed specifically in the paired ASI sensory neurons that are required for dauer bypass. ins-9 repression in dauer-constitutive mutants requires DPY-21, SET-4 and the FoxO transcription factor DAF-16, which is the main target of DAF-2 ILS. By contrast, autosomal genes encoding major agonist ILPs that promote reproductive development are not repressed by DPY-21, SET-4 or DAF-16/FoxO. Our results implicate SET-4 as a sensory rheostat that reinforces developmental fates in response to environmental cues by modulating autocrine and paracrine DAF-2 ILS.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/growth & development , Environment , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Neuronal Plasticity , Sensory Receptor Cells/physiology , Animals , Caenorhabditis elegans/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, X-Linked , Larva/metabolism , Male , Models, Biological , Mutation/genetics , Sex Characteristics , Transcriptome/genetics
5.
Cell Rep ; 17(5): 1227-1237, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783938

ABSTRACT

Vitamin D has multiple roles, including the regulation of bone and calcium homeostasis. Deficiency of 25-hydroxyvitamin D, the major circulating form of vitamin D, is associated with an increased risk of age-related chronic diseases, including Alzheimer's disease, Parkinson's disease, cognitive impairment, and cancer. In this study, we utilized Caenorhabditis elegans to examine the mechanism by which vitamin D influences aging. We found that vitamin-D3-induced lifespan extension requires the stress response pathway genes skn-1, ire-1, and xbp-1. Vitamin D3 (D3) induced expression of SKN-1 target genes but not canonical targets of XBP-1. D3 suppressed an important molecular pathology of aging, that of widespread protein insolubility, and prevented toxicity caused by human ß-amyloid. Our observation that D3 improves protein homeostasis and slows aging highlights the importance of maintaining appropriate vitamin D serum levels and may explain why such a wide variety of human age-related diseases are associated with vitamin D deficiency.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Homeostasis/drug effects , Longevity/physiology , Protein Serine-Threonine Kinases/genetics , Stress, Physiological/genetics , Transcription Factors/genetics , Vitamin D/pharmacology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Calcitriol/metabolism , Carrier Proteins/metabolism , Cholecalciferol/metabolism , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Protein Aggregates , Protein Serine-Threonine Kinases/metabolism , Solubility , Transcription Factors/metabolism , Unfolded Protein Response/drug effects
6.
G3 (Bethesda) ; 6(6): 1751-6, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27172199

ABSTRACT

Metazoan introns contain a polypyrimidine tract immediately upstream of the AG dinucleotide that defines the 3' splice site. In the nematode Caenorhabditis elegans, 3' splice sites are characterized by a highly conserved UUUUCAG/R octamer motif. While the conservation of pyrimidines in this motif is strongly suggestive of their importance in pre-mRNA splicing, in vivo evidence in support of this is lacking. In an N-ethyl-N-nitrosourea (ENU) mutagenesis screen in Caenorhabditis elegans, we have isolated a strain containing a point mutation in the octamer motif of a 3' splice site in the daf-12 gene. This mutation, a single base T-to-G transversion at the -5 position relative to the splice site, causes a strong daf-12 loss-of-function phenotype by abrogating splicing. The resulting transcript is predicted to encode a truncated DAF-12 protein generated by translation into the retained intron, which contains an in-frame stop codon. Other than the perfectly conserved AG dinucleotide at the site of splicing, G at the -5 position of the octamer motif is the most uncommon base in C. elegans 3' splice sites, occurring at closely paired sites where the better match to the splicing consensus is a few bases downstream. Our results highlight both the biological importance of the highly conserved -5 uridine residue in the C. elegans 3' splice site octamer motif as well as the utility of using ENU as a mutagen to study the function of polypyrimidine tracts and other AU- or AT-rich motifs in vivo.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Ethylnitrosourea/toxicity , Introns , Mutagenesis/drug effects , RNA Splice Sites , Animals , Base Sequence , Caenorhabditis elegans/metabolism , Chromosome Mapping , Mutation , Nucleotide Motifs , Phenotype , Polymorphism, Single Nucleotide , Protein Binding , RNA Splicing Factors/metabolism , Sequence Deletion
7.
G3 (Bethesda) ; 6(2): 351-6, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26628482

ABSTRACT

Chromoanasynthesis is a recently discovered phenomenon in humans with congenital diseases that is characterized by complex genomic rearrangements (CGRs) resulting from aberrant repair of catastrophic chromosomal damage. How these CGRs are induced is not known. Here, we describe the structure and function of dpDp667, a causative CGR that emerged from a Caenorhabditis elegans dauer suppressor screen in which animals were treated with the point mutagen N-ethyl-N-nitrosourea (ENU). dpDp667 comprises nearly 3 Mb of sequence on the right arm of the X chromosome, contains three duplications and one triplication, and is devoid of deletions. Sequences from three out of the four breakpoint junctions in dpDp667 reveal microhomologies that are hallmarks of chromoanasynthetic CGRs. Our findings suggest that environmental insults and physiological processes that cause point mutations may give rise to chromoanasynthetic rearrangements associated with congenital disease. The relatively subtle phenotype of animals harboring dpDp667 suggests that the prevalence of CGRs in the genomes of mutant and/or phenotypically unremarkable animals may be grossly underestimated.


Subject(s)
Caenorhabditis elegans/genetics , Gene Rearrangement , Genome, Helminth , Genomics , Mutagenesis , Animals , Caenorhabditis elegans/drug effects , Chromosome Breakpoints , Ethylnitrosourea/toxicity , Gene Dosage , Genomics/methods , Mutagenesis/drug effects , Sequence Analysis, DNA , X Chromosome
8.
Cell Metab ; 19(1): 73-83, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24411940

ABSTRACT

Small-molecule ligands of nuclear hormone receptors (NHRs) govern the transcriptional regulation of metazoan development, cell differentiation, and metabolism. However, the physiological ligands of many NHRs remain poorly characterized, primarily due to lack of robust analytical techniques. Using comparative metabolomics, we identified endogenous steroids that act as ligands of the C. elegans NHR, DAF-12, a vitamin D and liver X receptor homolog regulating larval development, fat metabolism, and lifespan. The identified molecules feature unexpected chemical modifications and include only one of two DAF-12 ligands reported earlier, necessitating a revision of previously proposed ligand biosynthetic pathways. We further show that ligand profiles are regulated by a complex enzymatic network, including the Rieske oxygenase DAF-36, the short-chain dehydrogenase DHS-16, and the hydroxysteroid dehydrogenase HSD-1. Our results demonstrate the advantages of comparative metabolomics over traditional candidate-based approaches and provide a blueprint for the identification of ligands for other C. elegans and mammalian NHRs.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Longevity/physiology , Metabolomics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cholestenes/chemistry , Cholestenes/metabolism , Gas Chromatography-Mass Spectrometry , Ligands , Magnetic Resonance Spectroscopy , Mutation/genetics , Organ Specificity , Signal Transduction , Steroids/metabolism
9.
Aging Cell ; 12(5): 932-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23786484

ABSTRACT

The AGC family serine-threonine kinases Akt and Sgk are similar in primary amino acid sequence and in vitro substrate specificity, and both kinases are thought to directly phosphorylate and inhibit FoxO transcription factors. In the nematode Caenorhabditis elegans, it is well established that AKT-1 controls dauer arrest and lifespan by regulating the subcellular localization of the FoxO transcription factor DAF-16. SGK-1 is thought to act similarly to AKT-1 in lifespan control by phosphorylating and inhibiting the nuclear translocation of DAF-16/FoxO. Using sgk-1 null and gain-of-function mutants, we now provide multiple lines of evidence indicating that AKT-1 and SGK-1 influence C. elegans lifespan, stress resistance, and DAF-16/FoxO activity in fundamentally different ways. Whereas AKT-1 shortens lifespan, SGK-1 promotes longevity in a DAF-16-/FoxO-dependent manner. In contrast to AKT-1, which reduces resistance to multiple stresses, SGK-1 promotes resistance to oxidative stress and ultraviolet radiation but inhibits thermotolerance. Analysis of several DAF-16/FoxO target genes that are repressed by AKT-1 reveals that SGK-1 represses a subset of these genes while having little influence on the expression of others. Accordingly, unlike AKT-1, which promotes the cytoplasmic sequestration of DAF-16/FoxO, SGK-1 does not influence DAF-16/FoxO subcellular localization. Thus, in spite of their similar in vitro substrate specificities, Akt and Sgk influence longevity, stress resistance, and FoxO activity through distinct mechanisms in vivo. Our findings highlight the need for a re-evaluation of current paradigms of FoxO regulation by Sgk.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Longevity/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Female , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Male , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism
10.
Genetics ; 194(3): 619-29, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23733789

ABSTRACT

During embryogenesis, an essential process known as dosage compensation is initiated to equalize gene expression from sex chromosomes. Although much is known about how dosage compensation is established, the consequences of modulating the stability of dosage compensation postembryonically are not known. Here we define a role for the Caenorhabditis elegans dosage compensation complex (DCC) in the regulation of DAF-2 insulin-like signaling. In a screen for dauer regulatory genes that control the activity of the FoxO transcription factor DAF-16, we isolated three mutant alleles of dpy-21, which encodes a conserved DCC component. Knockdown of multiple DCC components in hermaphrodite and male animals indicates that the dauer suppression phenotype of dpy-21 mutants is due to a defect in dosage compensation per se. In dpy-21 mutants, expression of several X-linked genes that promote dauer bypass is elevated, including four genes encoding components of the DAF-2 insulin-like pathway that antagonize DAF-16/FoxO activity. Accordingly, dpy-21 mutation reduced the expression of DAF-16/FoxO target genes by promoting the exclusion of DAF-16/FoxO from nuclei. Thus, dosage compensation enhances dauer arrest by repressing X-linked genes that promote reproductive development through the inhibition of DAF-16/FoxO nuclear translocation. This work is the first to establish a specific postembryonic function for dosage compensation in any organism. The influence of dosage compensation on dauer arrest, a larval developmental fate governed by the integration of multiple environmental inputs and signaling outputs, suggests that the dosage compensation machinery may respond to external cues by modulating signaling pathways through chromosome-wide regulation of gene expression.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/growth & development , Dosage Compensation, Genetic , Embryonic Development , Insulin/metabolism , Transcription Factors/genetics , Animals , Caenorhabditis elegans/genetics , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Genes, X-Linked , Insulin/genetics , Male , Mutation , Receptor, Insulin/genetics , Signal Transduction
11.
G3 (Bethesda) ; 3(5): 841-50, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23550118

ABSTRACT

Sterol-sensing nuclear receptors and insulin-like growth factor signaling play evolutionarily conserved roles in the control of aging. In the nematode Caenorhabditis elegans, bile acid-like steroid hormones known as dafachronic acids (DAs) influence longevity by binding to and regulating the activity of the conserved nuclear receptor DAF-12, and the insulin receptor (InsR) ortholog DAF-2 controls life span by inhibiting the FoxO transcription factor DAF-16. How the DA/DAF-12 pathway interacts with DAF-2/InsR signaling to control life span is poorly understood. Here we specifically investigated the roles of liganded and unliganded DAF-12 in life span control in the context of reduced DAF-2/InsR signaling. In animals with reduced daf-2/InsR activity, mutations that either reduce DA biosynthesis or fully abrogate DAF-12 activity shorten life span, suggesting that liganded DAF-12 promotes longevity. In animals with reduced DAF-2/InsR activity induced by daf-2/InsR RNAi, both liganded and unliganded DAF-12 promote longevity. However, in daf-2/InsR mutants, liganded and unliganded DAF-12 act in opposition to control life span. Thus, multiple DAF-12 activities influence life span in distinct ways in contexts of reduced DAF-2/InsR signaling. Our findings establish new roles for a conserved steroid signaling pathway in life span control and elucidate interactions among DA biosynthetic pathways, DAF-12, and DAF-2/InsR signaling in aging.


Subject(s)
Caenorhabditis elegans/physiology , Hormones/pharmacology , Insulin/metabolism , Longevity/drug effects , Signal Transduction/drug effects , Steroids/pharmacology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Cholestenes/metabolism , Genes, Helminth , Germ Cells/drug effects , Germ Cells/metabolism , Ligands , Models, Biological , Mutation/genetics , Receptor, Insulin/metabolism , Transcription, Genetic/drug effects
12.
PLoS Genet ; 9(12): e1004020, 2013.
Article in English | MEDLINE | ID: mdl-24385923

ABSTRACT

Recent work has identified changes in the metabolism of the aromatic amino acid tyrosine as a risk factor for diabetes and a contributor to the development of liver cancer. While these findings could suggest a role for tyrosine as a direct regulator of the behavior of cells and tissues, evidence for this model is currently lacking. Through the use of RNAi and genetic mutants, we identify tatn-1, which is the worm ortholog of tyrosine aminotransferase and catalyzes the first step of the conserved tyrosine degradation pathway, as a novel regulator of the dauer decision and modulator of the daf-2 insulin/IGF-1-like (IGFR) signaling pathway in Caenorhabditis elegans. Mutations affecting tatn-1 elevate tyrosine levels in the animal, and enhance the effects of mutations in genes that lie within the daf-2/insulin signaling pathway or are otherwise upstream of daf-16/FOXO on both dauer formation and worm longevity. These effects are mediated by elevated tyrosine levels as supplemental dietary tyrosine mimics the phenotypes produced by a tatn-1 mutation, and the effects still occur when the enzymes needed to convert tyrosine into catecholamine neurotransmitters are missing. The effects on dauer formation and lifespan require the aak-2/AMPK gene, and tatn-1 mutations increase phospho-AAK-2 levels. In contrast, the daf-16/FOXO transcription factor is only partially required for the effects on dauer formation and not required for increased longevity. We also find that the controlled metabolism of tyrosine by tatn-1 may function normally in dauer formation because the expression of the TATN-1 protein is regulated both by daf-2/IGFR signaling and also by the same dietary and environmental cues which influence dauer formation. Our findings point to a novel role for tyrosine as a developmental regulator and modulator of longevity, and support a model where elevated tyrosine levels play a causal role in the development of diabetes and cancer in people.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/growth & development , Longevity/genetics , Metabolic Networks and Pathways/genetics , Tyrosine Transaminase/genetics , Tyrosine/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , Humans , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Mutation , RNA Interference , Receptor, Insulin/metabolism , Transcription Factors/genetics , Tyrosine/metabolism
13.
Aging (Albany NY) ; 2(10): 742-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20975207

ABSTRACT

FoxO transcription factors (TFs) extend lifespan in invertebrates and may participate in the control of human longevity. The role of FoxO TFs in lifespan regulation has been studied most extensively inC. elegans, where a conserved insulin/insulin-like growth factor signaling (IIS) pathway and the germline both control lifespan by regulating the subcellular localization of the FoxO transcription factor DAF-16. Although the control of FoxO activity through modulation of its subcellular localization is well established, nuclear translocation of FoxO is not sufficient for full FoxO activation, suggesting that undiscovered inputs regulate FoxO activity after its translocation to the nucleus. We have recently discovered a new conserved pathway, the EAK (enhancer-of-akt-1) pathway, which acts in parallel to the Akt/PKB family of serine-threonine kinases to regulate DAF-16/FoxO activity. Whereas mutation of Akt/PKB promotes the nuclear accumulation of DAF-16/FoxO, mutation of eak genes increases nuclear DAF-16/FoxO activity without influencing DAF-16/FoxO subcellular localization. Thus, EAK proteins regulate the activity of nuclear DAF-16/FoxO. Two EAK proteins, EAK-2/HSD-1 and EAK-7, influence C. elegans lifespan and are conserved in mammals. The discovery of the EAK pathway defines a new conserved FoxO regulatory input and may have implications relevant to aging and the pathogenesis of aging-associated diseases.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Longevity/physiology , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Active Transport, Cell Nucleus/physiology , Aging/physiology , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Disease/etiology , Forkhead Transcription Factors/metabolism , Humans , Mice , Models, Biological , Osteoporosis/etiology , Osteoporosis/metabolism , Oxygenases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
14.
Cell Metab ; 12(1): 30-41, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20620993

ABSTRACT

FoxO transcription factors control development and longevity in diverse species. Although FoxO regulation via changes in its subcellular localization is well established, little is known about how FoxO activity is regulated in the nucleus. Here, we show that the conserved C. elegans protein EAK-7 acts in parallel to the serine/threonine kinase AKT-1 to inhibit the FoxO transcription factor DAF-16. Loss of EAK-7 activity promotes diapause and longevity in a DAF-16/FoxO-dependent manner. Whereas akt-1 mutation activates DAF-16/FoxO by promoting its translocation from the cytoplasm to the nucleus, eak-7 mutation increases nuclear DAF-16/FoxO activity without influencing DAF-16/FoxO subcellular localization. Thus, EAK-7 and AKT-1 inhibit DAF-16/FoxO activity via distinct mechanisms. Our results implicate EAK-7 as a FoxO regulator and highlight the biological impact of a regulatory pathway that governs the activity of nuclear FoxO without altering its subcellular location.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Longevity , Mutation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
15.
Dev Biol ; 340(2): 605-12, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20178781

ABSTRACT

Steroid hormone and insulin/insulin-like growth factor signaling (IIS) pathways control development and lifespan in the nematode Caenorhabditis elegans by regulating the activity of the nuclear receptor DAF-12 and the FoxO transcription factor DAF-16, respectively. The DAF-12 ligands Delta(4)- and Delta(7)-dafachronic acid (DA) promote bypass of the dauer diapause and proper gonadal migration during larval development; in adults, DAs influence lifespan. Whether Delta(4)- and Delta(7)-DA have unique biological functions is not known. We identified the 3-beta-hydroxysteroid dehydrogenase (3betaHSD) family member HSD-1, which participates in Delta(4)-DA biosynthesis, as an inhibitor of DAF-16/FoxO activity. Whereas IIS promotes the cytoplasmic sequestration of DAF-16/FoxO, HSD-1 inhibits nuclear DAF-16/FoxO activity without affecting DAF-16/FoxO subcellular localization. Thus, HSD-1 and IIS inhibit DAF-16/FoxO activity via distinct and complementary mechanisms. In adults, HSD-1 was required for full lifespan extension in IIS mutants, indicating that HSD-1 interactions with IIS are context-dependent. In contrast to the Delta(7)-DA biosynthetic enzyme DAF-36, HSD-1 is dispensable for proper gonadal migration and lifespan extension induced by germline ablation. These findings provide insights into the molecular interface between DA and IIS pathways and suggest that Delta(4)- and Delta(7)-DA pathways have unique as well as overlapping biological functions in the control of development and lifespan.


Subject(s)
Caenorhabditis elegans/physiology , Cholestenes/metabolism , Gene Expression Regulation, Developmental , Longevity/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Genes, Helminth , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Longevity/genetics , Models, Biological , Mutation , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...