Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
G3 (Bethesda) ; 9(10): 3477-3488, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31444297

ABSTRACT

To understand the genetic basis of complex traits, it is important to be able to efficiently phenotype many genetically distinct individuals. In the nematode Caenorhabditis elegans, individuals have been isolated from diverse populations around the globe and whole-genome sequenced. As a result, hundreds of wild strains with known genome sequences can be used for genome-wide association studies (GWAS). However, phenotypic analysis of these strains can be laborious, particularly for quantitative traits requiring multiple measurements per strain. Starvation resistance is likely a fitness-proximal trait for nematodes, and it is related to metabolic disease risk in humans. However, natural variation in C. elegans starvation resistance has not been systematically characterized, and precise measurement of the trait is time-intensive. Here, we developed a population-selection-and-sequencing-based approach to phenotype starvation resistance in a pool of 96 wild strains. We used restriction site-associated DNA sequencing (RAD-seq) to infer the frequency of each strain among survivors in a mixed culture over time during starvation. We used manual starvation survival assays to validate the trait data, confirming that strains that increased in frequency over time are starvation-resistant relative to strains that decreased in frequency. Further, we found that variation in starvation resistance is significantly associated with variation at a region on chromosome III. Using a near-isogenic line (NIL), we showed the importance of this genomic interval for starvation resistance. This study demonstrates the feasibility of using population selection and sequencing in an animal model for phenotypic analysis of quantitative traits, documents natural variation of starvation resistance in C. elegans, and identifies a genomic region that contributes to such variation.


Subject(s)
Adaptation, Biological/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Chromosome Mapping , Chromosomes , Selection, Genetic , Starvation , Animals , Genome-Wide Association Study , Phenotype , Quantitative Trait, Heritable
2.
Curr Biol ; 29(14): 2380-2388.e5, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31280992

ABSTRACT

The roundworm C. elegans reversibly arrests larval development during starvation [1], but extended early-life starvation reduces reproductive success [2, 3]. Maternal dietary restriction (DR) buffers progeny from starvation as young larvae, preserving reproductive success [4]. However, the developmental basis of reduced fertility following early-life starvation is unknown, and it is unclear how maternal diet modifies developmental physiology in progeny. We show here that extended starvation in first-stage (L1) larvae followed by unrestricted feeding results in a variety of developmental abnormalities in the reproductive system, including proliferative germ-cell tumors and uterine masses that express neuronal and epidermal cell fate markers. We found that maternal DR and reduced maternal insulin/insulin-like growth factor (IGF) signaling (IIS) increase oocyte provisioning of vitellogenin lipoprotein, reducing penetrance of starvation-induced abnormalities in progeny, including tumors. Furthermore, we show that maternal DR and reduced maternal IIS reduce IIS in progeny. daf-16/FoxO and skn-1/Nrf, transcriptional effectors of IIS, are required in progeny for maternal DR and increased vitellogenin provisioning to suppress starvation-induced abnormalities. daf-16/FoxO activity in somatic tissues is sufficient to suppress starvation-induced abnormalities, suggesting cell-nonautonomous regulation of reproductive system development. This work reveals that early-life starvation compromises reproductive development and that vitellogenin-mediated intergenerational insulin/IGF-to-insulin/IGF signaling mediates adaptation to nutrient availability.


Subject(s)
Adaptation, Physiological , Caenorhabditis elegans/physiology , Signal Transduction , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , Insulin/metabolism , Larva/genetics , Larva/growth & development , Larva/physiology , Nutrients/physiology , Somatomedins/metabolism , Vitellogenins/metabolism
3.
PLoS Genet ; 15(5): e1008056, 2019 05.
Article in English | MEDLINE | ID: mdl-31086367

ABSTRACT

The six C. elegans vulval precursor cells (VPCs) are induced to form the 3°-3°-2°-1°-2°-3° pattern of cell fates with high fidelity. In response to EGF signal, the LET-60/Ras-LIN-45/Raf-MEK-2/MEK-MPK-1/ERK canonical MAP kinase cascade is necessary to induce 1° fate and synthesis of DSL ligands for the lateral Notch signal. In turn, LIN-12/Notch receptor is necessary to induce neighboring cells to become 2°. We previously showed that, in response to graded EGF signal, the modulatory LET-60/Ras-RGL-1/RalGEF-RAL-1/Ral signal promotes 2° fate in support of LIN-12. In this study, we identify two key differences between RGL-1 and RAL-1. First, deletion of RGL-1 confers no overt developmental defects, while previous studies showed RAL-1 to be essential for viability and fertility. From this observation, we hypothesize that the essential functions of RAL-1 are independent of upstream activation. Second, RGL-1 plays opposing and genetically separable roles in VPC fate patterning. RGL-1 promotes 2° fate via canonical GEF-dependent activation of RAL-1. Conversely, RGL-1 promotes 1° fate via a non-canonical GEF-independent activity. Our genetic epistasis experiments are consistent with RGL-1 functioning in the modulatory 1°-promoting AGE-1/PI3-Kinase-PDK-1-AKT-1 cascade. Additionally, animals lacking RGL-1 experience 15-fold higher rates of VPC patterning errors compared to the wild type. Yet VPC patterning in RGL-1 deletion mutants is not more sensitive to environmental perturbations. We propose that RGL-1 functions to orchestrate opposing 1°- and 2°-promoting modulatory cascades to decrease developmental stochasticity. We speculate that such switches are broadly conserved but mostly masked by paralog redundancy or essential functions.


Subject(s)
Caenorhabditis elegans/genetics , Epidermal Growth Factor/genetics , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/genetics , Vulva/metabolism , Animals , Body Patterning/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Epidermal Growth Factor/metabolism , Epistasis, Genetic , Female , Fertility/genetics , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Vulva/cytology , Vulva/growth & development , raf Kinases/genetics , raf Kinases/metabolism , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , ras Proteins/genetics , ras Proteins/metabolism
4.
Genetics ; 211(1): 349-361, 2019 01.
Article in English | MEDLINE | ID: mdl-30425043

ABSTRACT

The Caenorhabditis elegans insulin-like signaling network supports homeostasis and developmental plasticity. The genome encodes 40 insulin-like peptides and one known receptor. Feedback regulation has been reported, but the extent of feedback and its effect on signaling dynamics in response to changes in nutrient availability has not been determined. We measured messenger RNA expression for each insulin-like peptide, the receptor daf-2, components of the PI3K pathway, and its transcriptional effectors daf-16/FoxO and skn-1/Nrf at high temporal resolution during transition from a starved, quiescent state to a fed, growing state in wild type and mutants affecting daf-2/InsR and daf-16/FoxO. We also analyzed the effect of temperature on insulin-like gene expression. We found that most PI3K pathway components and insulin-like peptides are affected by signaling activity, revealing pervasive positive and negative feedback regulation at intra- and intercellular levels. Reporter gene analysis demonstrated that the daf-2/InsR agonist daf-28 positively regulates its own transcription and that the putative agonist ins-6 cross-regulates DAF-28 protein expression through feedback. Our results show that positive and negative feedback regulation of insulin-like signaling is widespread, giving rise to an organismal FoxO-to-FoxO signaling network that supports homeostasis during fluctuations in nutrient availability.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Feedback, Physiological , Receptor, Insulin/metabolism , Signal Transduction , Somatomedins/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Receptor, Insulin/genetics , Somatomedins/genetics
5.
Nat Commun ; 9(1): 5152, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514845

ABSTRACT

Insulin and insulin-like signaling regulates a broad spectrum of growth and metabolic responses to a variety of internal and environmental stimuli. For example, the inhibition of insulin-like signaling in C. elegans mediates its response to both osmotic stress and starvation. We report that in response to osmotic stress the cytosolic sulfotransferase SSU-1 antagonizes insulin-like signaling and promotes developmental arrest. Both SSU-1 and the DAF-16 FOXO transcription factor, which is activated when insulin signaling is low, are needed to drive specific responses to reduced insulin-like signaling. We demonstrate that SSU-1 functions in a single pair of sensory neurons to control intercellular signaling via the nuclear hormone receptor NHR-1 and promote both the specific transcriptional response to osmotic stress and altered lysophosphatidylcholine metabolism. Our results show the requirement of a sulfotransferase-nuclear hormone receptor neurohormonal signaling pathway for some but not all consequences of reduced insulin-like signaling.


Subject(s)
Caenorhabditis elegans/metabolism , Nerve Tissue Proteins/drug effects , Neurotransmitter Agents/metabolism , Receptor, Insulin/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfotransferases/antagonists & inhibitors , Animals , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cloning, Molecular , Embryo, Nonmammalian , Embryonic Development/genetics , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Insulin/metabolism , Lysophosphatidylcholines/metabolism , Mutagenesis , Osmotic Pressure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sensory Receptor Cells/drug effects , Starvation , Stress, Physiological , Sulfotransferases/genetics , Sulfotransferases/metabolism
6.
BMC Biol ; 16(1): 112, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30296941

ABSTRACT

BACKGROUND: Developmental physiology is very sensitive to nutrient availability. For instance, in the nematode Caenorhabditis elegans, newly hatched L1-stage larvae require food to initiate postembryonic development. In addition, larvae arrested in the dauer diapause, a non-feeding state of developmental arrest that occurs during the L3 stage, initiate recovery when exposed to food. Despite the essential role of food in C. elegans development, the contribution of food perception versus ingestion on physiology has not been delineated. RESULTS: We used a pharmacological approach to uncouple the effects of food (bacteria) perception and ingestion in C. elegans. Perception was not sufficient to promote postembryonic development in L1-stage larvae. However, L1 larvae exposed to food without ingestion failed to develop upon return to normal culture conditions, instead displaying an irreversible arrest phenotype. Inhibition of gene expression during perception rescued subsequent development, demonstrating that the response to perception without feeding is deleterious. Perception altered DAF-16/FOXO subcellular localization, reflecting activation of insulin/IGF signaling (IIS). The insulin-like peptide daf-28 was specifically required, suggesting perception in chemosensory neurons, where it is expressed, regulates peptide synthesis and possibly secretion. However, genetic manipulation of IIS did not modify the irreversible arrest phenotype caused by food perception, revealing that wild-type function of the IIS pathway is not required to produce this phenotype and that other pathways affected by perception of food in the absence of its ingestion are likely to be involved. Gene expression and Nile red staining showed that food perception could alter lipid metabolism and storage. We found that starved larvae sense environmental polypeptides, with similar molecular and developmental effects as perception of bacteria. Environmental polypeptides also promoted recovery from dauer diapause, suggesting that perception of polypeptides plays an important role in the life history of free-living nematodes. CONCLUSIONS: We conclude that actual ingestion of food is required to initiate postembryonic development in C. elegans. We also conclude that polypeptides are perceived as a food-associated cue in this and likely other animals, initiating a signaling and gene regulatory cascade that alters metabolism in anticipation of feeding and development, but that this response is detrimental if feeding does not occur.


Subject(s)
Caenorhabditis elegans/drug effects , Eating/drug effects , Gene Expression/drug effects , Ivermectin/administration & dosage , Peptides/physiology , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cues , Food , Insulin/metabolism , Larva/drug effects , Larva/growth & development , Larva/metabolism , Signal Transduction/drug effects , Somatomedins/metabolism
7.
Cell Rep ; 24(10): 2669-2681.e5, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184501

ABSTRACT

C. elegans vulval precursor cell (VPC) fates are patterned by an epidermal growth factor (EGF) gradient. High-dose EGF induces 1° VPC fate, and lower dose EGF contributes to 2° fate in support of LIN-12/Notch. We previously showed that the EGF 2°-promoting signal is mediated by LET-60/Ras switching effectors, from the canonical Raf-MEK-ERK mitogen-activated protein (MAP) kinase cascade that promotes 1° fate to the non-canonical RalGEF-Ral that promotes 2° fate. Of oncogenic Ras effectors, RalGEF-Ral is by far the least well understood. We use genetic analysis to identify an effector cascade downstream of C. elegans RAL-1/Ral, starting with an established Ral binding partner, Exo84 of the exocyst complex. Additionally, RAL-1 signals through GCK-2, a citron-N-terminal-homology-domain-containing MAP4 kinase, and PMK-1/p38 MAP kinase cascade to promote 2° fate. Our study delineates a Ral-dependent developmental signaling cascade in vivo, thus providing the mechanism by which lower EGF dose is transduced.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Microtubule-Associated Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ral GTP-Binding Proteins/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Microtubule-Associated Proteins/genetics , Models, Biological , Signal Transduction/genetics , Signal Transduction/physiology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , ral GTP-Binding Proteins/genetics
8.
Nat Cell Biol ; 19(3): 252-257, 2017 03.
Article in English | MEDLINE | ID: mdl-28166192

ABSTRACT

In 1893 August Weismann proposed that information about the environment could not pass from somatic cells to germ cells, a hypothesis now known as the Weismann barrier. However, recent studies have indicated that parental exposure to environmental stress can modify progeny physiology and that parental stress can contribute to progeny disorders. The mechanisms regulating these phenomena are poorly understood. We report that the nematode Caenorhabditis elegans can protect itself from osmotic stress by entering a state of arrested development and can protect its progeny from osmotic stress by increasing the expression of the glycerol biosynthetic enzyme GPDH-2 in progeny. Both of these protective mechanisms are regulated by insulin-like signalling: insulin-like signalling to the intestine regulates developmental arrest, while insulin-like signalling to the maternal germline regulates glycerol metabolism in progeny. Thus, there is a heritable link between insulin-like signalling to the maternal germline and progeny metabolism and gene expression. We speculate that analogous modulation of insulin-like signalling to the germline is responsible for effects of the maternal environment on human diseases that involve insulin signalling, such as obesity and type-2 diabetes.


Subject(s)
Caenorhabditis elegans/physiology , Germ Cells/metabolism , Insulin/metabolism , Osmotic Pressure , Signal Transduction , Stress, Physiological , Animals , Bacterial Infections/pathology , Caenorhabditis elegans/microbiology , Intestines/embryology , MAP Kinase Signaling System , Starvation
9.
Worm ; 5(2): e1175196, 2016.
Article in English | MEDLINE | ID: mdl-27383290

ABSTRACT

Post-embryonic development is governed by nutrient availability. L1 arrest, dauer formation and aging illustrate how starvation, anticipation of starvation and caloric restriction have profound influence on C. elegans development, respectively. Insulin-like signaling through the Forkhead box O transcription factor daf-16/FoxO regulates each of these processes. We recently reported that ins-4, ins-6 and daf-28 promote L1 development from the intestine and chemosensory neurons, similar to their role in dauer development. daf-16 functions cell-nonautonomously in regulation of L1 arrest, dauer development and aging. Discrepancies in daf-16 sites of action have been reported in each context, but the consensus implicates epidermis, intestine and nervous system. We suggest technical limitations of the experimental approach responsible for discrepant results. Steroid hormone signaling through daf-12/NHR is known to function downstream of daf-16 in control of dauer development, but signaling pathways mediating cell-nonautonomous effects of daf-16 in aging and L1 arrest had not been identified. We recently showed that daf-16 promotes L1 arrest by inhibiting daf-12/NHR and dbl-1/TGF-ß Sma/Mab signaling, two pathways that promote L1 development in fed larvae. We will review these results on L1 arrest and speculate on why there are so many signals and signaling centers regulating post-embryonic development.

10.
PLoS Genet ; 11(12): e1005731, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26656736

ABSTRACT

Nutrient availability has profound influence on development. In the nematode C. elegans, nutrient availability governs post-embryonic development. L1-stage larvae remain in a state of developmental arrest after hatching until they feed. This "L1 arrest" (or "L1 diapause") is associated with increased stress resistance, supporting starvation survival. Loss of the transcription factor daf-16/FOXO, an effector of insulin/IGF signaling, results in arrest-defective and starvation-sensitive phenotypes. We show that daf-16/FOXO regulates L1 arrest cell-nonautonomously, suggesting that insulin/IGF signaling regulates at least one additional signaling pathway. We used mRNA-seq to identify candidate signaling molecules affected by daf-16/FOXO during L1 arrest. dbl-1/TGF-ß, a ligand for the Sma/Mab pathway, daf-12/NHR and daf-36/oxygenase, an upstream component of the daf-12 steroid hormone signaling pathway, were up-regulated during L1 arrest in a daf-16/FOXO mutant. Using genetic epistasis analysis, we show that dbl-1/TGF-ß and daf-12/NHR steroid hormone signaling pathways are required for the daf-16/FOXO arrest-defective phenotype, suggesting that daf-16/FOXO represses dbl-1/TGF-ß, daf-12/NHR and daf-36/oxygenase. The dbl-1/TGF-ß and daf-12/NHR pathways have not previously been shown to affect L1 development, but we found that disruption of these pathways delayed L1 development in fed larvae, consistent with these pathways promoting development in starved daf-16/FOXO mutants. Though the dbl-1/TGF-ß and daf-12/NHR pathways are epistatic to daf-16/FOXO for the arrest-defective phenotype, disruption of these pathways does not suppress starvation sensitivity of daf-16/FOXO mutants. This observation uncouples starvation survival from developmental arrest, indicating that DAF-16/FOXO targets distinct effectors for each phenotype and revealing that inappropriate development during starvation does not cause the early demise of daf-16/FOXO mutants. Overall, this study shows that daf-16/FOXO promotes developmental arrest cell-nonautonomously by repressing pathways that promote larval development.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Embryonic Development/genetics , Forkhead Transcription Factors/genetics , Insulin/genetics , Neuropeptides/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Somatomedins/genetics , Transforming Growth Factor beta/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/biosynthesis , Embryo, Nonmammalian , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Developmental , Humans , Insulin/metabolism , Larva/genetics , Larva/growth & development , Neuropeptides/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Signal Transduction , Somatomedins/metabolism , Starvation , Transforming Growth Factor beta/biosynthesis
11.
Mol Cell ; 53(2): 209-20, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24389102

ABSTRACT

Diverse environmental cues converge on and are integrated by the mTOR signaling network to control cellular growth and homeostasis. The mammalian Tsc1-Tsc2 GTPase activating protein (GAP) heterodimer is a critical negative regulator of Rheb and mTOR activation. The RalGAPα-RalGAPß heterodimer shares sequence and structural similarity with Tsc1-Tsc2. Unexpectedly, we observed that C. elegans expresses orthologs for the Rheb and RalA/B GTPases and for RalGAPα/ß, but not Tsc1/2. This prompted our investigation to determine whether RalGAPs additionally modulate mTOR signaling. We determined that C. elegans RalGAP loss decreased lifespan, consistent with a Tsc-like function. Additionally, RalGAP suppression in mammalian cells caused RalB-selective activation and Sec5- and exocyst-dependent engagement of mTORC1 and suppression of autophagy. Unexpectedly, we also found that Tsc1-Tsc2 loss activated RalA/B independently of Rheb-mTOR signaling. Finally, RalGAP suppression caused mTORC1-dependent pancreatic tumor cell invasion. Our findings identify an unexpected crosstalk and integration of the Ral and mTOR signaling networks.


Subject(s)
Autophagy/genetics , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/cytology , Cellular Senescence/genetics , GTP Phosphohydrolases/metabolism , Monomeric GTP-Binding Proteins/physiology , Neoplasm Invasiveness/genetics , TOR Serine-Threonine Kinases/metabolism , ral GTP-Binding Proteins/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Ras Homolog Enriched in Brain Protein , Signal Transduction , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...