Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Zebrafish ; 11(2): 85-97, 2014 Apr.
Article En | MEDLINE | ID: mdl-24428354

Transgenic expression of bacterial nitroreductase (NTR) facilitates chemically-inducible targeted cell ablation. In zebrafish, the NTR system enables studies of cell function and cellular regeneration. Metronidazole (MTZ) has become the most commonly used prodrug substrate for eliciting cell loss in NTR-expressing transgenic zebrafish due to the cell-specific nature of its cytotoxic derivatives. Unfortunately, MTZ treatments required for effective cell ablation border toxic effects, and, thus, likely incur undesirable nonspecific effects. Here, we tested whether a triple mutant variant of NTR, previously shown to display improved activity in bacterial assays, can solve this issue by promoting cell ablation in zebrafish using reduced prodrug treatment regimens. We generated several complementary transgenic zebrafish lines expressing either wild-type or mutant NTR (mutNTR) in specific neural cell types, and assayed prodrug-induced cell ablation kinetics using confocal time series imaging and plate reader-based quantification of fluorescent reporters expressed in targeted cell types. The results show that cell ablation can be achieved in mutNTR expressing transgenic lines with markedly shortened prodrug exposure times and/or at lower prodrug concentrations. The mutNTR variant characterized here can circumvent problematic nonspecific/toxic effects arising from low prodrug conversion efficiency, thus increasing the effectiveness and versatility of this selective cell ablation methodology.


Ablation Techniques/methods , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Metronidazole/metabolism , Neurons/physiology , Nitroreductases/genetics , Prodrugs/metabolism , Regeneration/drug effects , Zebrafish/physiology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/physiology , Cell Death/drug effects , Dose-Response Relationship, Drug , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Larva/genetics , Larva/physiology , Metronidazole/pharmacology , Nitroreductases/metabolism , Prodrugs/pharmacology , Time Factors , Zebrafish/genetics , Zebrafish/growth & development
2.
BMC Biol ; 10: 93, 2012 Nov 30.
Article En | MEDLINE | ID: mdl-23198762

BACKGROUND: We have investigated a simple strategy for enhancing transgene expression specificity by leveraging genetic silencer elements. The approach serves to restrict transgene expression to a tissue of interest - the nervous system in the example provided here - thereby promoting specific/exclusive targeting of discrete cellular subtypes. Recent innovations are bringing us closer to understanding how the brain is organized, how neural circuits function, and how neurons can be regenerated. Fluorescent proteins enable mapping of the 'connectome', optogenetic tools allow excitable cells to be short-circuited or hyperactivated, and targeted ablation of neuronal subtypes facilitates investigations of circuit function and neuronal regeneration. Optimally, such toolsets need to be expressed solely within the cell types of interest as off-site expression makes establishing causal relationships difficult. To address this, we have exploited a gene 'silencing' system that promotes neuronal specificity by repressing expression in non-neural tissues. This methodology solves non-specific background issues that plague large-scale enhancer trap efforts and may provide a means of leveraging promoters/enhancers that otherwise express too broadly to be of value for in vivo manipulations. RESULTS: We show that a conserved neuron-restrictive silencer element (NRSE) can function to restrict transgene expression to the nervous system. The neuron-restrictive silencing factor/repressor element 1 silencing transcription factor (NRSF/REST) transcriptional repressor binds NRSE/repressor element 1 (RE1) sites and silences gene expression in non-neuronal cells. Inserting NRSE sites into transgenes strongly biased expression to neural tissues. NRSE sequences were effective in restricting expression of bipartite Gal4-based 'driver' transgenes within the context of an enhancer trap and when associated with a defined promoter and enhancer. However, NRSE sequences did not serve to restrict expression of an upstream activating sequence (UAS)-based reporter/effector transgene when associated solely with the UAS element. Morpholino knockdown assays showed that NRSF/REST expression is required for NRSE-based transgene silencing. CONCLUSIONS: Our findings demonstrate that the addition of NRSE sequences to transgenes can provide useful new tools for functional studies of the nervous system. However, the general approach may be more broadly applicable; tissue-specific silencer elements are operable in tissues other than the nervous system, suggesting this approach can be similarly applied to other paradigms. Thus, creating synthetic associations between endogenous regulatory elements and tissue-specific silencers may facilitate targeting of cellular subtypes for which defined promoters/enhancers are lacking.


Neurons/metabolism , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified , Gene Expression Regulation/physiology , Gene Transfer Techniques , Genetic Engineering/methods , Plasmids , Promoter Regions, Genetic , Repressor Proteins/genetics , Transgenes , Zebrafish
3.
Future Med Chem ; 4(14): 1811-22, 2012 Sep.
Article En | MEDLINE | ID: mdl-23043478

Due to several inherent advantages, zebrafish are being utilized in increasingly sophisticated screens to assess the physiological effects of chemical compounds directly in living vertebrate organisms. Diverse screening platforms showcase these advantages. Morphological assays encompassing basic qualitative observations to automated imaging, manipulation, and data-processing systems provide whole organism to subcellular levels of detail. Behavioral screens extend chemical screening to the level of complex systems. In addition, zebrafish-based disease models provide a means of identifying new potential therapeutic strategies. Automated systems for handling/sorting, high-resolution imaging and quantitative data collection have significantly increased throughput in recent years. These advances will make it easier to capture multiple streams of information from a given sample and facilitate integration of zebrafish at the earliest stages of the drug-discovery process, providing potential solutions to current drug-development bottlenecks. Here we outline advances that have been made within the growing field of zebrafish chemical screening.


Drug Discovery/methods , Zebrafish/physiology , Animals , Behavior, Animal/drug effects , High-Throughput Screening Assays/methods , Luminescent Proteins/analysis , Zebrafish/anatomy & histology
4.
PLoS One ; 7(1): e29916, 2012.
Article En | MEDLINE | ID: mdl-22238673

Reporter-based assays underlie many high-throughput screening (HTS) platforms, but most are limited to in vitro applications. Here, we report a simple whole-organism HTS method for quantifying changes in reporter intensity in individual zebrafish over time termed, Automated Reporter Quantification in vivo (ARQiv). ARQiv differs from current "high-content" (e.g., confocal imaging-based) whole-organism screening technologies by providing a purely quantitative data acquisition approach that affords marked improvements in throughput. ARQiv uses a fluorescence microplate reader with specific detection functionalities necessary for robust quantification of reporter signals in vivo. This approach is: 1) Rapid; achieving true HTS capacities (i.e., >50,000 units per day), 2) Reproducible; attaining HTS-compatible assay quality (i.e., Z'-factors of ≥0.5), and 3) Flexible; amenable to nearly any reporter-based assay in zebrafish embryos, larvae, or juveniles. ARQiv is used here to quantify changes in: 1) Cell number; loss and regeneration of two different fluorescently tagged cell types (pancreatic beta cells and rod photoreceptors), 2) Cell signaling; relative activity of a transgenic Notch-signaling reporter, and 3) Cell metabolism; accumulation of reactive oxygen species. In summary, ARQiv is a versatile and readily accessible approach facilitating evaluation of genetic and/or chemical manipulations in living zebrafish that complements current "high-content" whole-organism screening methods by providing a first-tier in vivo HTS drug discovery platform.


Gene Dosage , Genes, Reporter , High-Throughput Screening Assays/methods , Image Processing, Computer-Assisted/methods , Zebrafish , Animals , Animals, Genetically Modified , Automation/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Embryo, Nonmammalian , Gene Dosage/physiology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Osmolar Concentration , Reproducibility of Results , Validation Studies as Topic , Zebrafish/embryology , Zebrafish/genetics
5.
Methods Mol Biol ; 571: 151-66, 2009.
Article En | MEDLINE | ID: mdl-19763965

Zebrafish have emerged as a powerful model organism to study neutrophil chemotaxis and inflammation in vivo. Studies of neutrophil chemotaxis in animal models have previously been hampered both by the limited number of specimens available for analysis and by the need for invasive procedures to perform intravital microscopy. Due to the transparency and cell permeability of zebrafish embryos these limitations are circumvented, and the zebrafish system is amenable to both live time-lapse imaging of neutrophil chemotaxis and for screening of the effects of chemical compounds on the inflammatory response in vivo. Here, we describe methods to analyze neutrophil-directed migration toward wounds using both fixed embryos by myeloperoxidase activity assay, and live embryos by time-lapse microscopy. Further, methods are described for the evaluation of the effects of chemical compounds on neutrophil motility and the innate immune responses in zebrafish embryos.


Neutrophils/cytology , Animals , Chemotaxis, Leukocyte/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Peroxidase/metabolism , Zebrafish , Zebrafish Proteins/metabolism
6.
Development ; 136(15): 2591-600, 2009 Aug.
Article En | MEDLINE | ID: mdl-19570844

Epidermal hyperproliferation and inflammation are hallmarks of the human condition psoriasis. Here, we report that a zebrafish line with a mutation in the cargo adaptor protein Clint1 exhibits psoriasis-like phenotypes including epithelial hyperproliferation and leukocyte infiltration. Clint1 is an ENTH domain-containing protein that binds SNARE proteins and functions in vesicle trafficking; however, its in vivo function in animal models has not been reported to date. The clint1 mutants exhibit chronic inflammation characterized by increased Interleukin 1beta expression, leukocyte infiltration, bidirectional trafficking and phagocytosis of cellular debris. The defects in clint1 mutants can be rescued by expression of zebrafish clint1 and can be phenocopied with clint1-specific morpholinos, supporting an essential role for Clint1 in epidermal development. Interaction studies suggest that Clint1 and Lethal giant larvae 2 function synergistically to regulate epidermal homeostasis. Accordingly, clint1 mutants show impaired hemidesmosome formation, loss of cell-cell contacts and increased motility suggestive of epithelial to mesenchymal transition. Taken together, our findings describe a novel function for the ENTH domain protein Clint1 in epidermal development and inflammation and suggest that its deficiency in zebrafish generates a phenotype that resembles the human condition psoriasis.


Epidermis/metabolism , Homeostasis , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Death , Cell Proliferation , Desmosomes/metabolism , Desmosomes/ultrastructure , Epidermis/pathology , Epidermis/ultrastructure , Epithelium/metabolism , Epithelium/ultrastructure , Gene Expression Regulation, Developmental , Inflammation/pathology , Leukocytes/cytology , Leukocytes/metabolism , Mesoderm/metabolism , Mesoderm/ultrastructure , Mutagenesis, Insertional , Mutation/genetics , Phagocytosis , Phenotype , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , beta Karyopherins/metabolism
7.
Dev Comp Immunol ; 33(11): 1212-7, 2009 Nov.
Article En | MEDLINE | ID: mdl-19619578

Zebrafish have emerged as a powerful model system to study leukocyte recruitment and inflammation. Here we characterize the morphology and function of inflammatory macrophages in zebrafish larvae. These macrophages can be distinguished from neutrophils by immunolabeling of L-Plastin without MPO co-expression and by an elongated morphology. Live imaging of transgenic zMPO:GFP larvae demonstrate that GFP(lo) macrophages migrate to wounds by extension of thin pseudopods and carry out phagocytosis of tissue debris, and FACS analysis of leukocyte markers indicates expression of CSF1R in these macrophages. These findings identify distinct functional and morphological characteristics of inflammatory macrophages in zebrafish larvae.


Antigens, Differentiation/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Zebrafish/immunology , Animals , Animals, Genetically Modified , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Cell Movement/immunology , Cell Separation , Flow Cytometry , Inflammation , Larva/immunology , Macrophages/immunology , Macrophages/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Peroxidase/metabolism , Phagocytosis , Pseudopodia/immunology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/immunology
8.
Cell Microbiol ; 11(5): 755-68, 2009 May.
Article En | MEDLINE | ID: mdl-19207728

Pseudomonas aeruginosa is an opportunistic human pathogen that can cause serious infection in those with deficient or impaired phagocytes. We have developed the optically transparent and genetically tractable zebrafish embryo as a model for systemic P. aeruginosa infection. Despite lacking adaptive immunity at this developmental stage, zebrafish embryos were highly resistant to P. aeruginosa infection, but as in humans, phagocyte depletion dramatically increased their susceptibility. The virulence of an attenuated P. aeruginosa strain lacking a functional Type III secretion system was restored upon phagocyte depletion, suggesting that this system influences virulence through its effects on phagocytes. Intravital imaging revealed bacterial interactions with multiple blood cell types. Neutrophils and macrophages rapidly phagocytosed and killed P. aeruginosa, suggesting that both cell types play a role in protection against infection. Intravascular aggregation of erythrocytes and other blood cells with resultant circulatory blockage was observed immediately upon infection, which may be relevant to the pathogenesis of thrombotic complications of human P. aeruginosa infections. The real-time visualization capabilities and genetic tractability of the zebrafish infection model should enable elucidation of molecular and cellular details of P. aeruginosa pathogenesis in conditions associated with neutropenia or impaired phagocyte function.


Bacterial Secretion Systems/physiology , Embryo, Nonmammalian/microbiology , Phagocytes/physiology , Pseudomonas aeruginosa/pathogenicity , Zebrafish/microbiology , Animals , Bacterial Secretion Systems/genetics , Embryo, Nonmammalian/cytology , Immunity, Innate , Macrophages/cytology , Macrophages/microbiology , Macrophages/physiology , Models, Animal , Neutrophils/cytology , Neutrophils/microbiology , Neutrophils/physiology , Phagocytes/cytology , Phagocytes/microbiology , Phagocytosis , Pseudomonas aeruginosa/genetics , Virulence , Zebrafish/embryology
9.
Dev Dyn ; 238(1): 86-99, 2009 Jan.
Article En | MEDLINE | ID: mdl-19097072

Factor for adipocyte differentiation 24 (fad24) is a novel gene that has been implicated in adipocyte differentiation and DNA replication. In a screen for zebrafish mutants that have an abnormal tissue distribution of neutrophils, we identified an insertional allele of fad24, fad24hi1019. Homozygous fad24hi1019 larvae exhibit muscle degeneration accompanied by leukocyte infiltration. Muscle degeneration was extensive and included tissue apoptosis and disorganized, poorly striated muscle fibers. Blocking apoptosis using pan-caspase inhibitors resulted in decreased neutrophil recruitment into the body of the larva, suggesting a causative link between apoptosis and leukocyte infiltration. These findings suggest that zebrafish is a powerful genetic model system to address the interplay between muscle degeneration and leukocyte infiltration, and indicate that tissue apoptosis may contribute to neutrophil recruitment in some inflammatory states.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Leukocytes/metabolism , Muscular Atrophy , Mutation , Nuclear Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Cell Nucleus/metabolism , Humans , In Situ Hybridization , Lipid Metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nuclear Proteins/genetics , Oligonucleotides, Antisense/metabolism , Zebrafish/anatomy & histology , Zebrafish/physiology , Zebrafish Proteins/genetics
10.
J Cell Sci ; 120(Pt 19): 3372-83, 2007 Oct 01.
Article En | MEDLINE | ID: mdl-17881499

The hallmark of chronic inflammation is the infiltration and persistence of leukocytes within inflamed tissue. Here, we describe the first zebrafish chronic inflammation mutant identified in an insertional mutagenesis screen for mutants that exhibit abnormal tissue distribution of neutrophils. We identified a mutant line with an insertion in the Hepatocyte growth factor activator inhibitor 1 gene (hai1; also known as Spint1) that showed accumulation of neutrophils in the fin. The mutant embryos exhibited inflammation in areas of epidermal hyperproliferation that was rescued by knock-down of the type II transmembrane serine protease Matriptase 1 (also known as St14), suggesting a novel role for Hai1-Matriptase 1 pathway in regulating inflammation. Using time-lapse microscopy of mutant embryos that express GFP from a neutrophil-specific promoter, we found that individual neutrophils in inflamed tissue displayed random motility characterized by periods of pausing alternating with periods of motility. During periods of persistent movement the cells were highly polarized, while the pausing modes were characterized by a loss of cell polarity. In contrast to responses to acute injury, neutrophils did not exhibit clear retrograde chemotaxis or resolution of inflammation in the mutant. These findings illustrate the utility of zebrafish as a new model system to study chronic inflammation and to visualize immune responses with high resolution in vivo.


Inflammation/pathology , Mutation , Proteinase Inhibitory Proteins, Secretory/genetics , Proteinase Inhibitory Proteins, Secretory/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish , Animals , Animals, Genetically Modified , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Movement/drug effects , Epithelium/anatomy & histology , Epithelium/physiology , Genes, Reporter , Humans , In Situ Hybridization , Inflammation/metabolism , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/physiology , Nitrobenzenes/pharmacology , Phenotype , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Sulfonamides/pharmacology , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/physiology
11.
J Leukoc Biol ; 80(6): 1281-8, 2006 Dec.
Article En | MEDLINE | ID: mdl-16963624

Neutrophil chemotaxis to sites of inflammation is a critical process during normal immune responses to tissue injury and infection and pathological immune responses leading to chronic inflammation. Although progress has been made in understanding the mechanisms that promote neutrophil recruitment to inflamed tissue, the mechanisms that regulate the resolution phase of the inflammatory response have remained relatively elusive. To define the mechanisms that regulate neutrophil-mediated inflammation in vivo, we have developed a novel transgenic zebrafish in which the neutrophils express GFP under control of the myeloperoxidase promoter (zMPO:GFP). Tissue injury induces a robust, inflammatory response, which is characterized by the rapid chemotaxis of neutrophils to the wound site. In vivo time-lapse imaging shows that neutrophils subsequently display directed retrograde chemotaxis back toward the vasculature. These findings implicate retrograde chemotaxis as a novel mechanism that regulates the resolution phase of the inflammatory response. The zMPO:GFP zebrafish provides unique insight into the mechanisms of neutrophil-mediated inflammation and thereby offers opportunities to identify new regulators of the inflammatory response in vivo.


Chemotaxis/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Wounds and Injuries/immunology , Zebrafish/immunology , Animals , Animals, Genetically Modified , Chemotaxis/genetics , Genetic Markers/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Microscopy, Fluorescence/methods , Microscopy, Video/methods , Neutrophil Infiltration/genetics , Neutrophils/pathology , Peroxidase/genetics , Peroxidase/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Wounds and Injuries/genetics , Wounds and Injuries/pathology , Zebrafish/genetics
12.
Nucleic Acids Res ; 32(22): 6469-78, 2004.
Article En | MEDLINE | ID: mdl-15598821

The HO gene in Saccharomyces cerevisiae is regulated by a large and complex promoter that is similar to promoters in higher order eukaryotes. Within this promoter are 10 potential binding sites for the a1-alpha2 heterodimer, which represses HO and other haploid-specific genes in diploid yeast cells. We have determined that a1-alpha2 binds to these sites with differing affinity, and that while certain strong-affinity sites are crucial for repression of HO, some of the weak-affinity sites are dispensable. However, these weak-affinity a1-alpha2-binding sites are strongly conserved in related yeast species and have a role in maintaining repression upon the loss of strong-affinity sites. We found that these weak sites are sufficient for a1-alpha2 to partially repress HO and recruit the Tup1-Cyc8 (Tup1-Ssn6) co-repressor complex to the HO promoter. We demonstrate that the Swi5 activator protein is not bound to URS1 in diploid cells, suggesting that recruitment of the Tup1-Cyc8 complex by a1-alpha2 prevents DNA binding by activator proteins resulting in repression of HO.


Deoxyribonucleases, Type II Site-Specific/genetics , Gene Expression Regulation, Fungal , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Base Sequence , Binding Sites , Cell Cycle , Chromatin Immunoprecipitation , Gene Silencing , Nuclear Proteins/metabolism , Phylogeny , Promoter Regions, Genetic , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/metabolism
13.
BMC Genomics ; 5(1): 59, 2004 Aug 26.
Article En | MEDLINE | ID: mdl-15331021

BACKGROUND: The analysis of gene expression using DNA microarrays provides genome wide profiles of the genes controlled by the presence or absence of a specific transcription factor. However, the question arises of whether a change in the level of transcription of a specific gene is caused by the transcription factor acting directly at the promoter of the gene or through regulation of other transcription factors working at the promoter. RESULTS: To address this problem we have devised a computational method that combines microarray expression and site preference data. We have tested this approach by identifying functional targets of the a1-alpha2 complex, which represses haploid-specific genes in the yeast Saccharomyces cerevisiae. Our analysis identified many known or suspected haploid-specific genes that are direct targets of the a1-alpha2 complex, as well as a number of previously uncharacterized targets. We were also able to identify a number of haploid-specific genes which do not appear to be direct targets of the a1-alpha2 complex, as well as a1-alpha2 target sites that do not repress transcription of nearby genes. Our method has a much lower false positive rate when compared to some of the conventional bioinformatic approaches. CONCLUSIONS: These findings show advantages of combining these two forms of data to investigate the mechanism of co-regulation of specific sets of genes.


Gene Expression Profiling/statistics & numerical data , Gene Expression Regulation, Fungal/genetics , Genome, Fungal , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Algorithms , Binding Sites/genetics , Chromosome Mapping/methods , Computational Biology/methods , DNA Mutational Analysis/statistics & numerical data , Diploidy , Haploidy , Mutation/genetics , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Predictive Value of Tests , Promoter Regions, Genetic/genetics , Software
14.
Structure ; 10(7): 961-71, 2002 Jul.
Article En | MEDLINE | ID: mdl-12121651

Triply mutated MATalpha2 protein, alpha2-3A, in which all three major groove-contacting residues are mutated to alanine, is defective in binding DNA alone or in complex with Mcm1 yet binds with MATa1 with near wild-type affinity and specificity. To gain insight into this unexpected behavior, we determined the crystal structure of the a1/alpha2-3A/DNA complex. The structure shows that the triple mutation causes a collapse of the alpha2-3A/DNA interface that results in a reorganized set of alpha2-3A/DNA contacts, thereby enabling the mutant protein to recognize the wild-type DNA sequence. Isothermal titration calorimetry measurements reveal that a much more favorable entropic component stabilizes the a1/alpha2-3A/DNA complex than the alpha2-3A/DNA complex. The combined structural and thermodynamic studies provide an explanation of how partner proteins influence the sequence specificity of a DNA binding protein.


Alanine/chemistry , DNA/chemistry , Homeodomain Proteins/chemistry , Repressor Proteins/chemistry , Calorimetry , Crystallography, X-Ray , Models, Molecular , Mutation , Nucleic Acid Conformation , Protein Conformation , Thermodynamics
15.
J Mol Biol ; 316(2): 247-56, 2002 Feb 15.
Article En | MEDLINE | ID: mdl-11851335

We have engineered enhanced DNA-binding function into the a1 homeodomain by making changes in a loop distant from the DNA-binding surface. Comparison of the free and bound a1 structures suggested a mechanism linking van der Waals stacking changes in this loop to the ordering of a final turn in the DNA-binding helix of a1. Inspection of the protein sequence revealed striking differences in amino acid identity at positions 24 and 25 compared to related homeodomain proteins. These positions lie in the loop connecting helix-1 and helix-2, which is involved in heterodimerization with the alpha 2 protein. A series of single and double amino acid substitutions (a1-Q24R, a1-S25Y, a1-S25F and a1-Q24R/S25Y) were engineered, expressed and purified for biochemical and biophysical study. Calorimetric measurements and HSQC NMR spectra confirm that the engineered variants are folded and are equally or more stable than the wild-type a1 homeodomain. NMR analysis of a1-Q24R/S25Y demonstrates that the DNA recognition helix (helix-3) is extended by at least one turn as a result of the changes in the loop connecting helix-1 and helix-2. As shown by EMSA, the engineered variants bind DNA with enhanced affinity (16-fold) in the absence of the alpha 2 cofactor and the variant alpha 2/a1 heterodimers bind cognate DNA with specificity and affinity reflective of the enhanced a1 binding affinity. Importantly, in vivo assays demonstrate that the a1-Q24R/S25Y protein binds with fivefold greater affinity than wild-type a1 and is able to partially suppress defects in repression by alpha 2 mutants. As a result of these studies, we show how subtle differences in residues at a surface distant from the functional site code for a conformational switch that allows the a1 homeodomain to become active in DNA binding in association with its cofactor alpha 2.


DNA/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Protein Engineering , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Base Sequence , Calorimetry , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , Dimerization , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Fungal , Homeodomain Proteins/genetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Precipitin Tests , Protein Binding , Protein Structure, Secondary , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Substrate Specificity
...