Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Methods ; 68(3): 529-35, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24990146

ABSTRACT

Recent breakthrough discoveries have shown that committed cell fates can be reprogrammed by genetic, chemical and environmental manipulations. The germline of the nematode Caenorhabditis elegans provides a tractable system for studying cell fate reprogramming within the context of a whole organism. To explore the possibility of using C. elegans in high-throughput screens (HTS), we developed a high-throughput workflow for testing compounds that modulate cell fate reprogramming. We utilized puf-8; lip-1 mutants that have enhanced MPK-1 (an ERK homolog)/MAP kinase (MAPK) signaling. Wild-type C. elegans hermaphrodites produce both sperm and oocytes, and are thus self-fertile. However, puf-8; lip-1 mutants produce only sperm and are sterile. Notably, compounds that pharmacologically down-regulate MPK-1 (an ERK homolog)/MAP kinase (MAPK) signaling are able to reprogram germ cell fate and restore fertility to these animals. puf-8; lip-1 mutants provide numerous challenges for HTS. First, they are sterile as homozygotes and must be maintained as heterozygotes using a balancer chromosome. Second, homozygous animals for experimentation must be physically separated from the rest of the population. Third, a high quality, high-content assay has not been developed to measure compound effects on germ cell fate reprogramming. Here we describe a semi-automated high-throughput workflow that enables effective sorting of homozygous puf-8; lip-1 mutants into 384-well plates using the COPAS™ BIOSORT. In addition, we have developed an image-based assay for rapidly measuring germ cell reprogramming by measuring the number of viable progeny in wells. The methods presented in this report enable the use of puf-8; lip-1 mutants in HTS campaigns for chemical modulators of germ cell reprogramming within the context of a whole organism.


Subject(s)
Caenorhabditis elegans/drug effects , Cellular Reprogramming/drug effects , Germ Cells/drug effects , Small Molecule Libraries/administration & dosage , Animals , Caenorhabditis elegans/growth & development , Cell Lineage/drug effects , High-Throughput Screening Assays , Male , Signal Transduction/drug effects
2.
Expert Opin Drug Discov ; 9(9): 1021-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24998976

ABSTRACT

INTRODUCTION: Many human diseases result from a failure of a single protein to achieve the correct folding and tertiary conformation. These so-called 'conformational diseases' involve diverse proteins and distinctive cellular pathologies. They all engage the proteostasis network (PN), to varying degrees in an attempt to mange cellular stress and restore protein homeostasis. The insulin/insulin-like growth factor signaling (IIS) pathway is a master regulator of cellular stress response, which is implicated in regulating components of the PN. AREAS COVERED: This review focuses on novel approaches to target conformational diseases. The authors discuss the evidence supporting the involvement of the IIS pathway in modulating the PN and regulating proteostasis in Caenorhabditis elegans. Furthermore, they review previous PN and IIS drug screens and explore the possibility of using C. elegans for whole organism-based drug discovery for modulators of IIS-proteostasis pathways. EXPERT OPINION: An alternative approach to develop individualized therapy for each conformational disease is to modulate the global PN. The involvement of the IIS pathway in regulating longevity and response to a variety of stresses is well documented. Increasing data now provide evidence for the close association between the IIS and the PN pathways. The authors believe that high-throughput screening campaigns, which target the C. elegans IIS pathway, may identify drugs that are efficacious in treating numerous conformational diseases.


Subject(s)
Caenorhabditis elegans/physiology , Drug Discovery/methods , Stress, Physiological/physiology , Animals , High-Throughput Screening Assays/methods , Humans , Insulin/physiology , Longevity/physiology , Protein Folding , Protein Structure, Tertiary/physiology , Signal Transduction/physiology , Somatomedins/physiology
3.
Hum Mol Genet ; 23(19): 5109-22, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24838286

ABSTRACT

The accumulation of serpin oligomers and polymers within the endoplasmic reticulum (ER) causes cellular injury in patients with the classical form α1-antitrypsin deficiency (ATD). To better understand the cellular and molecular genetic aspects of this disorder, we generated transgenic C. elegans strains expressing either the wild-type (ATM) or Z mutant form (ATZ) of the human serpin fused to GFP. Animals secreted ATM, but retained polymerized ATZ within dilated ER cisternae. These latter animals also showed slow growth, smaller brood sizes and decreased longevity; phenotypes observed in ATD patients or transgenic mouse lines expressing ATZ. Similar to mammalian models, ATZ was disposed of by autophagy and ER-associated degradation pathways. Mutant strains defective in insulin signaling (daf-2) also showed a marked decrease in ATZ accumulation. Enhanced ATZ turnover was associated with the activity of two proteins central to systemic/exogenous (exo)-RNAi pathway: the dsRNA importer, SID-1 and the argonaute, RDE-1. Animals with enhanced exo-RNAi activity (rrf-3 mutant) phenocopied the insulin signaling mutants and also showed increased ATZ turnover. Taken together, these studies allude to the existence of a novel proteostasis pathway that mechanistically links misfolded protein turnover to components of the systemic RNAi machinery.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , RNA Interference , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans Proteins/genetics , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum-Associated Degradation , Gene Expression , Genes, Reporter , Humans , Insulin/metabolism , Mice , Mice, Transgenic , Mutation , Phenotype , Promoter Regions, Genetic , Proteolysis , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serpins , Signal Transduction , Sodium-Hydrogen Exchangers/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/metabolism
4.
Hum Mol Genet ; 23(19): 5123-32, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24838285

ABSTRACT

α1-Antitrypsin deficiency (ATD) is a common genetic disorder that can lead to end-stage liver and lung disease. Although liver transplantation remains the only therapy currently available, manipulation of the proteostasis network (PN) by small molecule therapeutics offers great promise. To accelerate the drug-discovery process for this disease, we first developed a semi-automated high-throughput/content-genome-wide RNAi screen to identify PN modifiers affecting the accumulation of the α1-antitrypsin Z mutant (ATZ) in a Caenorhabditis elegans model of ATD. We identified 104 PN modifiers, and these genes were used in a computational strategy to identify human ortholog-ligand pairs. Based on rigorous selection criteria, we identified four FDA-approved drugs directed against four different PN targets that decreased the accumulation of ATZ in C. elegans. We also tested one of the compounds in a mammalian cell line with similar results. This methodology also proved useful in confirming drug targets in vivo, and predicting the success of combination therapy. We propose that small animal models of genetic disorders combined with genome-wide RNAi screening and computational methods can be used to rapidly, economically and strategically prime the preclinical discovery pipeline for rare and neglected diseases with limited therapeutic options.


Subject(s)
Drug Discovery , Genome-Wide Association Study , RNA Interference , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Animals , Caenorhabditis elegans , Computational Biology , Disease Models, Animal , Genomics , High-Throughput Screening Assays , Humans , Mutation , Protein Binding , Proteostasis Deficiencies/genetics , Reproducibility of Results , alpha 1-Antitrypsin Deficiency/drug therapy
5.
PLoS One ; 9(1): e87260, 2014.
Article in English | MEDLINE | ID: mdl-24498058

ABSTRACT

The classical form of α1-antitrypsin deficiency (ATD) is associated with hepatic fibrosis and hepatocellular carcinoma. It is caused by the proteotoxic effect of a mutant secretory protein that aberrantly accumulates in the endoplasmic reticulum of liver cells. Recently we developed a model of this deficiency in C. elegans and adapted it for high-content drug screening using an automated, image-based array scanning. Screening of the Library of Pharmacologically Active Compounds identified fluphenazine (Flu) among several other compounds as a drug which reduced intracellular accumulation of mutant α1-antitrypsin Z (ATZ). Because it is representative of the phenothiazine drug class that appears to have autophagy enhancer properties in addition to mood stabilizing activity, and can be relatively easily re-purposed, we further investigated its effects on mutant ATZ. The results indicate that Flu reverses the phenotypic effects of ATZ accumulation in the C. elegans model of ATD at doses which increase the number of autophagosomes in vivo. Furthermore, in nanomolar concentrations, Flu enhances the rate of intracellular degradation of ATZ and reduces the cellular ATZ load in mammalian cell line models. In the PiZ mouse model Flu reduces the accumulation of ATZ in the liver and mediates a decrease in hepatic fibrosis. These results show that Flu can reduce the proteotoxicity of ATZ accumulation in vivo and, because it has been used safely in humans, this drug can be moved rapidly into trials for liver disease due to ATD. The results also provide further validation for drug discovery using C. elegans models that can be adapted to high-content drug screening platforms and used together with mammalian cell line and animal models.


Subject(s)
Caenorhabditis elegans/metabolism , Disease Models, Animal , Fluphenazine/pharmacology , alpha 1-Antitrypsin Deficiency/prevention & control , Animals , Animals, Genetically Modified , Antipsychotic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , CHO Cells , Caenorhabditis elegans/genetics , Cricetinae , Cricetulus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hep G2 Cells , Humans , Immunoblotting , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Phagosomes/drug effects , Phagosomes/metabolism , Survival Analysis , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...