Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(25): eadg0506, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37352352

ABSTRACT

Gene activity defines cell identity, drives intercellular communication, and underlies the functioning of multicellular organisms. We present the single-cell resolution atlas of gene activity of a fertile adult metazoan: Caenorhabditis elegans. This compendium comprises 180 distinct cell types and 19,657 expressed genes. We predict 7541 transcription factor expression profile associations likely responsible for defining cellular identity. We predict thousands of intercellular interactions across the C. elegans body and the ligand-receptor pairs that mediate them, some of which we experimentally validate. We identify 172 genes that show consistent expression across cell types, are involved in basic and essential functions, and are conserved across phyla; therefore, we present them as experimentally validated housekeeping genes. We developed the WormSeq application to explore these data. In addition to the integrated gene-to-systems biology, we present genome-scale single-cell resolution testable hypotheses that we anticipate will advance our understanding of the molecular mechanisms, underlying the functioning of a multicellular organism and the perturbations that lead to its malfunction.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Transcription Factors/metabolism , Gene Expression Regulation , Gene Expression
2.
STAR Protoc ; 4(1): 101956, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36856764

ABSTRACT

A major barrier to immunostaining Caenorhabditis elegans is the permeabilization of the worm's cuticle without distorting or damaging its body. We present here a gel-based immobilization protocol for fixed worms coupled with chemical and enzymatic permeabilization. The permeabilization is followed by antibody staining and fluorescent imaging. This protocol can be modified for different fixatives, permeabilizing reagents, or molecular readouts. Unlike previous immunostaining approaches, such as freeze cracking or dissection, this protocol enables immunostaining across the whole body of a well-preserved C. elegans.


Subject(s)
Acrylic Resins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/chemistry , Fixatives , Antibodies
3.
Curr Biol ; 33(6): 1036-1046.e6, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36805847

ABSTRACT

Several molecules can extend healthspan and lifespan across organisms. However, most are upstream signaling hubs or transcription factors orchestrating complex anti-aging programs. Therefore, these molecules point to but do not reveal the fundamental mechanisms driving longevity. Instead, downstream effectors that are necessary and sufficient to promote longevity across conditions or organisms may reveal the fundamental anti-aging drivers. Toward this goal, we searched for effectors acting downstream of the transcription factor EB (TFEB), known as HLH-30 in C. elegans, because TFEB/HLH-30 is necessary across anti-aging interventions and its overexpression is sufficient to extend C. elegans lifespan and reduce biomarkers of aging in mammals including humans. As a result, we present an alcohol-dehydrogenase-mediated anti-aging response (AMAR) that is essential for C. elegans longevity driven by HLH-30 overexpression, caloric restriction, mTOR inhibition, and insulin-signaling deficiency. The sole overexpression of ADH-1 is sufficient to activate AMAR, which extends healthspan and lifespan by reducing the levels of glycerol-an age-associated and aging-promoting alcohol. Adh1 overexpression is also sufficient to promote longevity in yeast, and adh-1 orthologs are induced in calorically restricted mice and humans, hinting at ADH-1 acting as an anti-aging effector across phyla.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Humans , Animals , Mice , Longevity/physiology , Caenorhabditis elegans/genetics , Alcohol Dehydrogenase/genetics , Caenorhabditis elegans Proteins/genetics , Aging , Mammals , Basic Helix-Loop-Helix Transcription Factors
4.
PLoS Comput Biol ; 18(11): e1010715, 2022 11.
Article in English | MEDLINE | ID: mdl-36395331

ABSTRACT

Cell-cell interactions shape cellular function and ultimately organismal phenotype. Interacting cells can sense their mutual distance using combinations of ligand-receptor pairs, suggesting the existence of a spatial code, i.e., signals encoding spatial properties of cellular organization. However, this code driving and sustaining the spatial organization of cells remains to be elucidated. Here we present a computational framework to infer the spatial code underlying cell-cell interactions from the transcriptomes of the cell types across the whole body of a multicellular organism. As core of this framework, we introduce our tool cell2cell, which uses the coexpression of ligand-receptor pairs to compute the potential for intercellular interactions, and we test it across the Caenorhabditis elegans' body. Leveraging a 3D atlas of C. elegans' cells, we also implement a genetic algorithm to identify the ligand-receptor pairs most informative of the spatial organization of cells across the whole body. Validating the spatial code extracted with this strategy, the resulting intercellular distances are negatively correlated with the inferred cell-cell interactions. Furthermore, for selected cell-cell and ligand-receptor pairs, we experimentally confirm the communicatory behavior inferred with cell2cell and the genetic algorithm. Thus, our framework helps identify a code that predicts the spatial organization of cells across a whole-animal body.


Subject(s)
Caenorhabditis elegans , Cell Communication , Animals , Ligands , Communication , Phenotype
5.
PLoS Comput Biol ; 18(7): e1010295, 2022 07.
Article in English | MEDLINE | ID: mdl-35830477

ABSTRACT

The concept of "housekeeping gene" has been used for four decades but remains loosely defined. Housekeeping genes are commonly described as "essential for cellular existence regardless of their specific function in the tissue or organism", and "stably expressed irrespective of tissue type, developmental stage, cell cycle state, or external signal". However, experimental support for the tenet that gene essentiality is linked to stable expression across cell types, conditions, and organisms has been limited. Here we use genome-scale functional genomic screens together with bulk and single-cell sequencing technologies to test this link and optimize a quantitative and experimentally validated definition of housekeeping gene. Using the optimized definition, we identify, characterize, and provide as resources, housekeeping gene lists extracted from several human datasets, and 10 other animal species that include primates, chicken, and C. elegans. We find that stably expressed genes are not necessarily essential, and that the individual genes that are essential and stably expressed can considerably differ across organisms; yet the pathways enriched among these genes are conserved. Further, the level of conservation of housekeeping genes across the analyzed organisms captures their taxonomic groups, showing evolutionary relevance for our definition. Therefore, we present a quantitative and experimentally supported definition of housekeeping genes that can contribute to better understanding of their unique biological and evolutionary characteristics.


Subject(s)
Caenorhabditis elegans , Genes, Essential , Animals , Caenorhabditis elegans/genetics , Genes, Essential/genetics , Household Work , Humans
6.
PLoS Comput Biol ; 18(1): e1009776, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35007280

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pcbi.1007764.].

7.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607947

ABSTRACT

Plasticity in multicellular organisms involves signaling pathways converting contexts-either natural environmental challenges or laboratory perturbations-into context-specific changes in gene expression. Congruently, the interactions between the signaling molecules and transcription factors (TF) regulating these responses are also context specific. However, when a target gene responds across contexts, the upstream TF identified in one context is often inferred to regulate it across contexts. Reconciling these stable TF-target gene pair inferences with the context-specific nature of homeostatic responses is therefore needed. The induction of the Caenorhabditis elegans genes lipl-3 and lipl-4 is observed in many genetic contexts and is essential to survival during fasting. We find DAF-16/FOXO mediating lipl-4 induction in all contexts tested; hence, lipl-4 regulation seems context independent and compatible with across-context inferences. In contrast, DAF-16-mediated regulation of lipl-3 is context specific. DAF-16 reduces the induction of lipl-3 during fasting, yet it promotes it during oxidative stress. Through discrete dynamic modeling and genetic epistasis, we define that DAF-16 represses HLH-30/TFEB-the main TF activating lipl-3 during fasting. Contrastingly, DAF-16 activates the stress-responsive TF HSF-1 during oxidative stress, which promotes C. elegans survival through induction of lipl-3 Furthermore, the TF MXL-3 contributes to the dominance of HSF-1 at the expense of HLH-30 during oxidative stress but not during fasting. This study shows how context-specific diverting of functional interactions within a molecular network allows cells to specifically respond to a large number of contexts with a limited number of molecular players, a mode of transcriptional regulation we name "contextualized transcription."


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Fasting/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/genetics , Lipase/metabolism , Oxidative Stress/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Lipase/genetics , Lipolysis/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/physiology
8.
PLoS Genet ; 17(9): e1009736, 2021 09.
Article in English | MEDLINE | ID: mdl-34492009

ABSTRACT

Obesity and its associated metabolic syndrome are a leading cause of morbidity and mortality. Given the disease's heavy burden on patients and the healthcare system, there has been increased interest in identifying pharmacological targets for the treatment and prevention of obesity. Towards this end, genome-wide association studies (GWAS) have identified hundreds of human genetic variants associated with obesity. The next challenge is to experimentally define which of these variants are causally linked to obesity, and could therefore become targets for the treatment or prevention of obesity. Here we employ high-throughput in vivo RNAi screening to test for causality 293 C. elegans orthologs of human obesity-candidate genes reported in GWAS. We RNAi screened these 293 genes in C. elegans subject to two different feeding regimens: (1) regular diet, and (2) high-fructose diet, which we developed and present here as an invertebrate model of diet-induced obesity (DIO). We report 14 genes that promote obesity and 3 genes that prevent DIO when silenced in C. elegans. Further, we show that knock-down of the 3 DIO genes not only prevents excessive fat accumulation in primary and ectopic fat depots but also improves the health and extends the lifespan of C. elegans overconsuming fructose. Importantly, the direction of the association between expression variants in these loci and obesity in mice and humans matches the phenotypic outcome of the loss-of-function of the C. elegans ortholog genes, supporting the notion that some of these genes would be causally linked to obesity across phylogeny. Therefore, in addition to defining causality for several genes so far merely correlated with obesity, this study demonstrates the value of model systems compatible with in vivo high-throughput genetic screening to causally link GWAS gene candidates to human diseases.


Subject(s)
Caenorhabditis elegans/genetics , Genetic Predisposition to Disease , Obesity/genetics , Animals , Dietary Carbohydrates/administration & dosage , Fructose/administration & dosage , Gene Expression , Homeostasis , Humans , Meta-Analysis as Topic , Phenotype
9.
PLoS Comput Biol ; 16(5): e1007764, 2020 05.
Article in English | MEDLINE | ID: mdl-32396573

ABSTRACT

Diverse algorithms can integrate transcriptomics with genome-scale metabolic models (GEMs) to build context-specific metabolic models. These algorithms require identification of a list of high confidence (core) reactions from transcriptomics, but parameters related to identification of core reactions, such as thresholding of expression profiles, can significantly change model content. Importantly, current thresholding approaches are burdened with setting singular arbitrary thresholds for all genes; thus, resulting in removal of enzymes needed in small amounts and even many housekeeping genes. Here, we describe StanDep, a novel heuristic method for using transcriptomics to identify core reactions prior to building context-specific metabolic models. StanDep clusters gene expression data based on their expression pattern across different contexts and determines thresholds for each cluster using data-dependent statistics, specifically standard deviation and mean. To demonstrate the use of StanDep, we built hundreds of models for the NCI-60 cancer cell lines. These models successfully increased the inclusion of housekeeping reactions, which are often lost in models built using standard thresholding approaches. Further, StanDep also provided a transcriptomic explanation for inclusion of lowly expressed reactions that were otherwise only supported by model extraction methods. Our study also provides novel insights into how cells may deal with context-specific and ubiquitous functions. StanDep, as a MATLAB toolbox, is available at https://github.com/LewisLabUCSD/StanDep.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Metabolomics/methods , Algorithms , Genome , Humans , Metabolic Networks and Pathways , Models, Biological , Models, Theoretical , Transcriptome
10.
Nat Commun ; 11(1): 2587, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444616

ABSTRACT

The gut microbiota metabolizes drugs and alters their efficacy and toxicity. Diet alters drugs, the metabolism of the microbiota, and the host. However, whether diet-triggered metabolic changes in the microbiota can alter drug responses in the host has been largely unexplored. Here we show that dietary thymidine and serine enhance 5-fluoro 2'deoxyuridine (FUdR) toxicity in C. elegans through different microbial mechanisms. Thymidine promotes microbial conversion of the prodrug FUdR into toxic 5-fluorouridine-5'-monophosphate (FUMP), leading to enhanced host death associated with mitochondrial RNA and DNA depletion, and lethal activation of autophagy. By contrast, serine does not alter FUdR metabolism. Instead, serine alters E. coli's 1C-metabolism, reduces the provision of nucleotides to the host, and exacerbates DNA toxicity and host death without mitochondrial RNA or DNA depletion; moreover, autophagy promotes survival in this condition. This work implies that diet-microbe interactions can alter the host response to drugs without altering the drug or the host.


Subject(s)
Caenorhabditis elegans/drug effects , Floxuridine/toxicity , Food-Drug Interactions , Gastrointestinal Microbiome/drug effects , Serine/pharmacology , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Dietary Supplements , Escherichia coli/drug effects , Escherichia coli/metabolism , Floxuridine/pharmacokinetics , Folic Acid/metabolism , Gastrointestinal Microbiome/physiology , Thymidine/analogs & derivatives , Thymidine/metabolism , Thymidine/pharmacokinetics , Thymidine/pharmacology , Uracil Nucleotides/metabolism , Uracil Nucleotides/pharmacokinetics
11.
Methods Mol Biol ; 1787: 129-146, 2018.
Article in English | MEDLINE | ID: mdl-29736715

ABSTRACT

Caenorhabditis elegans is the first and only metazoan model that enables whole-body gene knockdown by simply feeding their standard laboratory diet, E. coli, carrying RNA interference (RNAi)-expressing constructs. The simplicity of the RNAi treatment, small size, and fast reproduction rate of C. elegans allow us to perform whole-animal high-throughput genetic screens in wild-type, mutant, or otherwise genetically modified C. elegans. In addition, more than 65% of C. elegans genes are conserved in mammals including human. In particular, C. elegans metabolic pathways are highly conserved, which supports the study of complex diseases such as obesity in this genetically tractable model system. In this chapter, we present a detailed protocol for automated high-throughput whole-animal RNAi screening to identify the pathways promoting obesity in diet-induced and genetically driven obese C. elegans. We describe an optimized high-content screening protocol to score fat mass and body fat distribution in whole animals at large scale. We provide optimized pipelines to automatically score phenotypes using the open-source CellProfiler platform within the context of supercomputer clusters. Further, we present a guideline to optimize information workflow from the automated microscope to a searchable database. The approaches described here enable unveiling the whole network of gene-gene and gene-environment interactions that define metabolic health or disease status in this proven model of human disease, but similar principles can be applied to other disease models.


Subject(s)
Gene Regulatory Networks , Obesity/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drug Discovery/methods , Drug Evaluation, Preclinical , Gene Regulatory Networks/drug effects , Genetic Predisposition to Disease , Humans , Molecular Imaging , Obesity/metabolism , RNA Interference
12.
Autophagy ; 12(4): 619-31, 2016.
Article in English | MEDLINE | ID: mdl-27050453

ABSTRACT

Lysosomes are highly acidic cellular organelles traditionally viewed as sacs of enzymes involved in digesting extracellular or intracellular macromolecules for the regeneration of basic building blocks, cellular housekeeping, or pathogen degradation. Bound by a single lipid bilayer, lysosomes receive their substrates by fusing with endosomes or autophagosomes, or through specialized translocation mechanisms such as chaperone-mediated autophagy or microautophagy. Lysosomes degrade their substrates using up to 60 different soluble hydrolases and release their products either to the cytosol through poorly defined exporting and efflux mechanisms or to the extracellular space by fusing with the plasma membrane. However, it is becoming evident that the role of the lysosome in nutrient homeostasis goes beyond the disposal of waste or the recycling of building blocks. The lysosome is emerging as a signaling hub that can integrate and relay external and internal nutritional information to promote cellular and organismal homeostasis, as well as a major contributor to the processing of energy-dense molecules like glycogen and triglycerides. Here we describe the current knowledge of the nutrient signaling pathways governing lysosomal function, the role of the lysosome in nutrient mobilization, and how lysosomes signal other organelles, distant tissues, and even themselves to ensure energy homeostasis in spite of fluctuations in energy intake. At the same time, we highlight the value of genomics approaches to the past and future discoveries of how the lysosome simultaneously executes and controls cellular homeostasis.


Subject(s)
Food , Homeostasis , Lysosomes/metabolism , Animals , Humans , Models, Biological , Signal Transduction
13.
Autophagy ; 11(1): 9-27, 2015.
Article in English | MEDLINE | ID: mdl-25569839

ABSTRACT

The cellular recycling process of autophagy has been extensively characterized with standard assays in yeast and mammalian cell lines. In multicellular organisms, numerous external and internal factors differentially affect autophagy activity in specific cell types throughout the stages of organismal ontogeny, adding complexity to the analysis of autophagy in these metazoans. Here we summarize currently available assays for monitoring the autophagic process in the nematode C. elegans. A combination of measuring levels of the lipidated Atg8 ortholog LGG-1, degradation of well-characterized autophagic substrates such as germline P granule components and the SQSTM1/p62 ortholog SQST-1, expression of autophagic genes and electron microscopy analysis of autophagic structures are presently the most informative, yet steady-state, approaches available to assess autophagy levels in C. elegans. We also review how altered autophagy activity affects a variety of biological processes in C. elegans such as L1 survival under starvation conditions, dauer formation, aging, and cell death, as well as neuronal cell specification. Taken together, C. elegans is emerging as a powerful model organism to monitor autophagy while evaluating important physiological roles for autophagy in key developmental events as well as during adulthood.


Subject(s)
Autophagy , Caenorhabditis elegans/cytology , Guidelines as Topic , Animals , Biological Assay , Caenorhabditis elegans/embryology , Embryonic Development , Models, Biological
14.
Methods ; 68(3): 492-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24784529

ABSTRACT

Fat accumulation is a complex phenotype affected by factors such as neuroendocrine signaling, feeding, activity, and reproductive output. Accordingly, the most informative screens for genes and compounds affecting fat accumulation would be those carried out in whole living animals. Caenorhabditis elegans is a well-established and effective model organism, especially for biological processes that involve organ systems and multicellular interactions, such as metabolism. Every cell in the transparent body of C. elegans is visible under a light microscope. Consequently, an accessible and reliable method to visualize worm lipid-droplet fat depots would make C. elegans the only metazoan in which genes affecting not only fat mass but also body fat distribution could be assessed at a genome-wide scale. Here we present a radical improvement in oil red O worm staining together with high-throughput image-based phenotyping. The three-step sample preparation method is robust, formaldehyde-free, and inexpensive, and requires only 15min of hands-on time to process a 96-well plate. Together with our free and user-friendly automated image analysis package, this method enables C. elegans sample preparation and phenotype scoring at a scale that is compatible with genome-wide screens. Thus we present a feasible approach to small-scale phenotyping and large-scale screening for genetic and/or chemical perturbations that lead to alterations in fat quantity and distribution in whole animals.


Subject(s)
Body Fat Distribution , Lipid Metabolism/genetics , Obesity/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Genome , High-Throughput Screening Assays , Models, Animal , Obesity/etiology , Obesity/genetics , Phenotype
15.
Nat Cell Biol ; 15(6): 668-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23604316

ABSTRACT

Fat is stored or mobilized according to food availability. Malfunction of the mechanisms that ensure this coordination underlie metabolic diseases in humans. In mammals, lysosomal and autophagic function is required for normal fat storage and mobilization in the presence or absence of food. Autophagy is tightly linked to nutrients. However, if and how lysosomal lipolysis is coupled to nutritional status remains to be determined. Here we identify MXL-3 and HLH-30 (TFEB orthologue) [corrected] as transcriptional switches coupling lysosomal lipolysis and autophagy to nutrient availability and controlling fat storage and ageing in Caenorhabditis elegans. Transcriptional coupling of lysosomal lipolysis and autophagy to nutrients is also observed in mammals. Thus, MXL-3 and HLH-30 orchestrate an adaptive and conserved cellular response to nutritional status and regulate lifespan.


Subject(s)
Autophagy/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Lipolysis/physiology , Aging/genetics , Aging/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Fasting , Lipase/genetics , Lipase/metabolism , Lysosomes/metabolism , Trans-Activators/metabolism , Transcription Factors
16.
Genes Dev ; 27(4): 429-40, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23392608

ABSTRACT

Adaptation to nutrient scarcity depends on the activation of metabolic programs to efficiently use internal reserves of energy. Activation of these programs in abundant food regimens can extend life span. However, the common molecular and metabolic changes that promote adaptation to nutritional stress and extend life span are mostly unknown. Here we present a response to fasting, enrichment of ω-6 polyunsaturated fatty acids (PUFAs), which promotes starvation resistance and extends Caenorhabditis elegans life span. Upon fasting, C. elegans induces the expression of a lipase, which in turn leads to an enrichment of ω-6 PUFAs. Supplementing C. elegans culture media with these ω-6 PUFAs increases their resistance to starvation and extends their life span in conditions of food abundance. Supplementation of C. elegans or human epithelial cells with these ω-6 PUFAs activates autophagy, a cell recycling mechanism that promotes starvation survival and slows aging. Inactivation of C. elegans autophagy components reverses the increase in life span conferred by supplementing the C. elegans diet with these fasting-enriched ω-6 PUFAs. We propose that the salubrious effects of dietary supplementation with ω-3/6 PUFAs (fish oils) that have emerged from epidemiological studies in humans may be due to a similar activation of autophagic programs.


Subject(s)
Autophagy/physiology , Caenorhabditis elegans/physiology , Fatty Acids, Omega-6/metabolism , Longevity/physiology , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , Fasting/physiology , Gene Expression Regulation , HeLa Cells , Humans , Lipase/genetics , Lipase/metabolism , Lipolysis , Mice , Survival Analysis
17.
Nat Methods ; 9(7): 714-6, 2012 Apr 22.
Article in English | MEDLINE | ID: mdl-22522656

ABSTRACT

We present a toolbox for high-throughput screening of image-based Caenorhabditis elegans phenotypes. The image analysis algorithms measure morphological phenotypes in individual worms and are effective for a variety of assays and imaging systems. This WormToolbox is available through the open-source CellProfiler project and enables objective scoring of whole-worm high-throughput image-based assays of C. elegans for the study of diverse biological pathways that are relevant to human disease.


Subject(s)
Caenorhabditis elegans/cytology , High-Throughput Screening Assays , Image Processing, Computer-Assisted , Microscopy, Fluorescence/methods , Pattern Recognition, Automated/methods , Algorithms , Animals , High-Throughput Screening Assays/instrumentation , High-Throughput Screening Assays/methods , Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/instrumentation , Phenotype , Software
18.
Cell Metab ; 10(5): 430-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19883620

ABSTRACT

Genetic conservation allows ancient features of fat storage endocrine pathways to be explored in C. elegans. Multiple studies have used Nile red or BODIPY-labeled fatty acids to identify regulators of fat mass. When mixed with their food, E. coli bacteria, Nile red, and BODIPY-labeled fatty acids stain multiple spherical cellular structures in the C. elegans major fat storage organ, the intestine. However, here we demonstrate that, in the conditions previously reported, the lysosome-related organelles stained by Nile red and BODIPY-labeled fatty acids are not the C. elegans major fat storage compartment. We show that the major fat stores are contained in a distinct cellular compartment that is not stained by Nile red. Using biochemical assays, we validate oil red O staining as a method to assess major fat stores in C. elegans, allowing for efficient and accurate genetic and functional genomic screens for genes that control fat accumulation at the organismal level.


Subject(s)
Caenorhabditis elegans/metabolism , Cytoplasmic Vesicles/metabolism , Fats/metabolism , Animals , Azo Compounds , Boron Compounds , Caenorhabditis elegans/ultrastructure , Cytoplasmic Vesicles/ultrastructure , Fluorescent Dyes , Oxazines , Staining and Labeling
19.
Methods Mol Biol ; 486: 57-75, 2009.
Article in English | MEDLINE | ID: mdl-19347616

ABSTRACT

The nematode Caenorhabditis elegans shows a high degree of conservation of molecular pathways related to human disease, yet is only 1-mm long and can be considered as a microorganism. Because of the development of a simple but systematic RNA-interference (RNAi) methodology, C. elegans is the only metazoan in which the impact of "knocking-down" nearly every gene in the genome can be analyzed in a whole living animal. Both functional genomic studies and chemical screens can be carried out using C. elegans in vivo screens in a context that preserves intact cell-to-cell communication, neuroendocrine signaling, and every aspect of the animal's metabolism necessary to survive and reproduce in lab conditions. This feature enables studies that are impossible to undertake in cell-culture-based screens. Although genome-wide RNAi screens and limited small-molecule screens have been successfully performed in C. elegans, they are typically extremely labor-intensive. Furthermore, technical limitations have precluded quantitative measurements and time-resolved analyses.In this chapter, we provide detailed protocols to carry out automated high-throughput whole-animal RNAi and chemical screens. We describe methods to perform screens in solid and liquid media, in 96 and 384-well format, respectively. We describe the use of automated handling, sorting, and microscopy of worms. Finally, we give information about worm-adapted image analysis tools to quantify phenotypes. The technology presented here facilitates large-scale C. elegans genetic and chemical screens and it is expected to help shed light on relevant biological areas.


Subject(s)
Animals, Genetically Modified , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/genetics , RNA Interference/physiology , Animals , Gene Library , Survival Rate
20.
Science ; 322(5903): 957-60, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18988854

ABSTRACT

Fat metabolism, reproduction, and aging are intertwined regulatory axes; however, the mechanism by which they are coupled remains poorly understood. We found that germline stem cells (GSCs) actively modulate lipid hydrolysis in Caenorhabditis elegans, which in turn regulates longevity. GSC arrest promotes systemic lipolysis via induction of a specific fat lipase. Subsequently, fat mobilization is promoted and life span is prolonged. Constitutive expression of this lipase in fat storage tissue generates lean and long-lived animals. This lipase is a key factor in the lipid hydrolysis and increased longevity that are induced by decreased insulin signaling. These results suggest a link between C. elegans fat metabolism and longevity.


Subject(s)
Caenorhabditis elegans/physiology , Germ Cells/metabolism , Lipase/metabolism , Lipid Metabolism , Longevity , Stem Cells/metabolism , Aging , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation , Cell Proliferation , Forkhead Transcription Factors , Genes, Helminth , Germ Cells/cytology , Hydrolysis , Intestinal Mucosa/metabolism , Intestines/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Lipase/genetics , Models, Animal , Receptor, Insulin/metabolism , Reproduction , Signal Transduction , Stem Cells/cytology , Temperature , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...