Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Syst Biol ; 15(11): e9012, 2019 11.
Article in English | MEDLINE | ID: mdl-31777173

ABSTRACT

Size of organs/organisms is a polygenic trait. Many of the growth-regulatory genes constitute conserved growth signaling pathways. However, how these multiple genes are orchestrated at the systems level to attain the natural variation in size including sexual size dimorphism is mostly unknown. Here we take a multi-layered systems omics approach to study size variation in the Drosophila wing. We show that expression levels of many critical growth regulators such as Wnt and TGFß pathway components significantly differ between sexes but not between lines exhibiting size differences within each sex, suggesting a primary role of these regulators in sexual size dimorphism. Only a few growth genes including a receptor of steroid hormone ecdysone exhibit association with between-line size differences. In contrast, we find that between-line size variation is largely regulated by genes with a diverse range of cellular functions, most of which have never been implicated in growth. In addition, we show that expression quantitative trait loci (eQTLs) linked to these novel growth regulators accurately predict population-wide, between-line wing size variation. In summary, our study unveils differential gene regulatory systems that control wing size variation between and within sexes.


Subject(s)
Drosophila melanogaster/growth & development , Gene Expression Profiling/methods , Transforming Growth Factor beta/genetics , Wings, Animal/anatomy & histology , Wnt Proteins/genetics , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Male , Organ Size , Quantitative Trait Loci , Sequence Analysis, RNA , Sex Characteristics , Signal Transduction
2.
PLoS Genet ; 12(1): e1005616, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26751788

ABSTRACT

Organismal size depends on the interplay between genetic and environmental factors. Genome-wide association (GWA) analyses in humans have implied many genes in the control of height but suffer from the inability to control the environment. Genetic analyses in Drosophila have identified conserved signaling pathways controlling size; however, how these pathways control phenotypic diversity is unclear. We performed GWA of size traits using the Drosophila Genetic Reference Panel of inbred, sequenced lines. We find that the top associated variants differ between traits and sexes; do not map to canonical growth pathway genes, but can be linked to these by epistasis analysis; and are enriched for genes and putative enhancers. Performing GWA on well-studied developmental traits under controlled conditions expands our understanding of developmental processes underlying phenotypic diversity.


Subject(s)
Body Size/genetics , Drosophila melanogaster/genetics , Genetic Variation , Genome-Wide Association Study , Animals , Drosophila melanogaster/growth & development , Humans , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Signal Transduction
3.
Health Res Policy Syst ; 17(1): 10, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30691504

ABSTRACT

BACKGROUND: While the European Union is striving to become the 'Innovation Union', there remains a lack of quantifiable indicators to compare and benchmark regional innovation clusters. To address this issue, a HealthTIES (Healthcare, Technology and Innovation for Economic Success) consortium was funded by the European Union's Regions of Knowledge initiative, research and innovation funding programme FP7. HealthTIES examined whether the health technology innovation cycle was functioning differently in five European regional innovation clusters and proposed regional and joint actions to improve their performance. The clusters included BioCat (Barcelona, Catalonia, Spain), Medical Delta (Leiden, Rotterdam and Delft, South Holland, Netherlands), Oxford and Thames Valley (United Kingdom), Life Science Zürich (Switzerland), and Innova Észak-Alföld (Debrecen, Hungary). METHODS: Appreciation of the 'triple helix' of university-industry-government innovation provided the impetus for the development of two quantifiable innovation indexes and related indicators. The HealthTIES H-index is calculated for disease and technology platforms based on the h-index proposed by Hirsch. The HealthTIES Innovation Index is calculated for regions based on 32 relevant quantitative and discriminative indicators grouped into 12 categories and 3 innovation phases, namely 'Input' (n = 12), 'Innovation System' (n = 9) and 'Output' (n = 11). RESULTS: The HealthTIES regions had developed relatively similar disease and technology platform profiles, yet with distinctive strengths and weaknesses. The regional profiles of the innovation cycle in each of the three phases were surprisingly divergent. Comparative assessments based on the indicators and indexes helped identify and share best practice and inform regional and joint action plans to strengthen the competitiveness of the HealthTIES regions. CONCLUSION: The HealthTIES indicators and indexes provide useful practical tools for the measurement and benchmarking of university-industry-government innovation in European medical and life science clusters. They are validated internally within the HealthTIES consortium and appear to have a degree of external prima facie validity. Potentially, the tools and accompanying analyses can be used beyond the HealthTIES consortium to inform other regional governments, researchers and, possibly, large companies searching for their next location, analyse and benchmark 'triple helix' dynamics within their own networks over time, and to develop integrated public-private and cross-regional research and innovation strategies in Europe and beyond.


Subject(s)
Benchmarking , Biological Science Disciplines , Biomedical Research , Government , Industry , Universities , Biomedical Technology , Delivery of Health Care , Europe , European Union , Humans , Knowledge , Technology
4.
J Cell Sci ; 128(14): 2497-508, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26054799

ABSTRACT

The kinase TOR is found in two complexes, TORC1, which is involved in growth control, and TORC2, whose roles are less well defined. Here, we asked whether TORC2 has a role in sustaining cellular stress. We show that TORC2 inhibition in Drosophila melanogaster leads to a reduced tolerance to heat stress, whereas sensitivity to other stresses is not affected. Accordingly, we show that upon heat stress, both in the animal and Drosophila cultured S2 cells, TORC2 is activated and is required for maintaining the level of its known target, Akt1 (also known as PKB). We show that the phosphorylation of the stress-activated protein kinases is not modulated by TORC2 nor is the heat-induced upregulation of heat-shock proteins. Instead, we show, both in vivo and in cultured cells, that TORC2 is required for the assembly of heat-induced cytoprotective ribonucleoprotein particles, the pro-survival stress granules. These granules are formed in response to protein translation inhibition imposed by heat stress that appears to be less efficient in the absence of TORC2 function. We propose that TORC2 mediates heat resistance in Drosophila by promoting the cell autonomous formation of stress granules.


Subject(s)
Cytoplasmic Granules/metabolism , Drosophila Proteins/metabolism , Heat-Shock Response/physiology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cytoplasmic Granules/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics
5.
Genesis ; 54(11): 573-581, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27636555

ABSTRACT

The FLP/FRT system permits rapid phenotypic screening of homozygous lethal mutations in the context of a viable mosaic fly. Combining this system with ovoD dominant female-sterile transgenes enables efficient production of embryos derived from mutant germline clones lacking maternal contribution from a gene of interest. Two distinct sets of FRT chromosomes, carrying either the mini-white (w + mW.hs ), or rosy (ry+ ) and neomycin (neoR ) transgenes are in common use. Parallel ovoD lines were developed using w + mW.hs FRT insertions on the X and chromosomes 2R and 3L, as well as ry+ , neoR FRT insertions on 2L and 3R. Consequently, mutations isolated on the X, 2R and 3L chromosomes in a ry+ , neoR FRT background, are not amenable to germline clonal analysis without labor-intensive recombination onto chromosome arms containing a w + mW.hs FRT. Here we report the creation of a new ovoD line for the ry+ , neoR FRT insertion at position FRT42D on chromosome 2R, through induced recombination in males. To establish the developmental relevance of this reagent we characterized the maternal-effect phenotypes of novel brother of tout-velu alleles generated on an FRT42D chromosome in a somatic mosaic screen. We find that an apparent null mutation that causes severe defects in somatic tissues has a much milder effect on embryonic patterning, emphasizing the necessity of analyzing mutant phenotypes at multiple developmental stages.


Subject(s)
Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Infertility, Female/genetics , N-Acetylglucosaminyltransferases/genetics , Alleles , Animals , Drosophila melanogaster/growth & development , Female , Gene Expression Regulation, Developmental , Germ-Line Mutation/genetics , Humans , Male , Mosaicism/embryology , Phenotype , Synthetic Lethal Mutations/genetics , Transgenes
6.
PLoS Genet ; 9(7): e1003598, 2013.
Article in English | MEDLINE | ID: mdl-23874212

ABSTRACT

Appropriate expression of growth-regulatory genes is essential to ensure normal animal development and to prevent diseases like cancer. Gene regulation at the levels of transcription and translational initiation mediated by the Hippo and Insulin signaling pathways and by the TORC1 complex, respectively, has been well documented. Whether translational control mediated by RNA-binding proteins contributes to the regulation of cellular growth is less clear. Here, we identify Lingerer (Lig), an UBA domain-containing protein, as growth suppressor that associates with the RNA-binding proteins Fragile X mental retardation protein 1 (FMR1) and Caprin (Capr) and directly interacts with and regulates the RNA-binding protein Rasputin (Rin) in Drosophila melanogaster. lig mutant organs overgrow due to increased proliferation, and a reporter for the JAK/STAT signaling pathway is upregulated in a lig mutant situation. rin, Capr or FMR1 in combination as double mutants, but not the respective single mutants, display lig like phenotypes, implicating a redundant function of Rin, Capr and FMR1 in growth control in epithelial tissues. Thus, Lig regulates cell proliferation during development in concert with Rin, Capr and FMR1.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Fragile X Mental Retardation Protein/metabolism , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelium/growth & development , Epithelium/metabolism , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation, Developmental , Humans , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction
7.
Dev Biol ; 381(1): 97-106, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23773803

ABSTRACT

In Drosophila, growth takes place during the larval stages until the formation of the pupa. Starvation delays pupariation to allow prolonged feeding, ensuring that the animal reaches an appropriate size to form a fertile adult. Pupariation is induced by a peak of the steroid hormone ecdysone produced by the prothoracic gland (PG) after larvae have reached a certain body mass. Local downregulation of the insulin/insulin-like growth factor signaling (IIS) activity in the PG interferes with ecdysone production, indicating that IIS activity in the PG couples the nutritional state to development. However, the underlying mechanism is not well understood. In this study we show that the secreted Imaginal morphogenesis protein-Late 2 (Imp-L2), a growth inhibitor in Drosophila, is involved in this process. Imp-L2 inhibits the activity of the Drosophila insulin-like peptides by direct binding and is expressed by specific cells in the brain, the ring gland, the gut and the fat body. We demonstrate that Imp-L2 is required to regulate and adapt developmental timing to nutritional conditions by regulating IIS activity in the PG. Increasing Imp-L2 expression at its endogenous sites using an Imp-L2-Gal4 driver delays pupariation, while Imp-L2 mutants exhibit a slight acceleration of development. These effects are strongly enhanced by starvation and are accompanied by massive alterations of ecdysone production resulting most likely from increased Imp-L2 production by neurons directly contacting the PG and not from elevated Imp-L2 levels in the hemolymph. Taken together our results suggest that Imp-L2-expressing neurons sense the nutritional state of Drosophila larvae and coordinate dietary information and ecdysone production to adjust developmental timing under starvation conditions.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , RNA-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Ecdysone/metabolism , Ecdysterone/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Larva/growth & development , Mutation , Neurons/metabolism , Protein Isoforms , Signal Transduction , Transcription Factors/genetics , Transgenes
8.
PLoS Genet ; 7(8): e1002168, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21829386

ABSTRACT

The integrity of the intestinal epithelium is crucial for the barrier function of the gut. Replenishment of the gut epithelium by intestinal stem cells contributes to gut homeostasis, but how the differentiated enterocytes are protected against stressors is less well understood. Here we use the Drosophila larval hindgut as a model system in which damaged enterocytes are not replaced by stem cell descendants. By performing a thorough genetic analysis, we demonstrate that a signalling complex consisting of p38b and MK2 forms a branch of SAPK signalling that is required in the larval hindgut to prevent stress-dependent damage to the enterocytes. Impaired p38b/MK2 signalling leads to apoptosis of the enterocytes and a subsequent loss of hindgut epithelial integrity, as manifested by the deterioration of the overlaying muscle layer. Damaged hindguts show increased JNK activity, and removing upstream activators of JNK suppresses the loss of hindgut homeostasis. Thus, the p38/MK2 complex ensures homeostasis of the hindgut epithelium by counteracting JNK-mediated apoptosis of the enterocytes upon chronic stress.


Subject(s)
Apoptosis , Drosophila/enzymology , Enterocytes/enzymology , MAP Kinase Signaling System , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , Animals , Apoptosis/genetics , Drosophila/genetics , Female , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Mutation/genetics , Phosphorylation , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics
9.
Development ; 137(3): 499-506, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081194

ABSTRACT

Apical cell surfaces in metazoan epithelia, such as the wing disc of Drosophila, resemble polygons with different numbers of neighboring cells. The distribution of these polygon numbers has been shown to be conserved. Revealing the mechanisms that lead to this topology might yield insights into how the structural integrity of epithelial tissues is maintained. It has previously been proposed that cell division alone, or cell division in combination with cell rearrangements, is sufficient to explain the observed epithelial topology. Here, we extend this work by including an analysis of the clustering and the polygon distribution of mitotic cells. In addition, we study possible effects of cellular growth regulation by mechanical forces, as such regulation has been proposed to be involved in wing disc size regulation. We formulated several theoretical scenarios that differ with respect to whether cell rearrangements are allowed and whether cellular growth rates are dependent on mechanical stress. We then compared these scenarios with experimental data on the polygon distribution of the entire cell population, that of mitotic cells, as well as with data on mitotic clustering. Surprisingly, we observed considerably less clustering in our experiments than has been reported previously. Only scenarios that include mechanical-stress-dependent growth rates are in agreement with the experimental data. Interestingly, simulations of these scenarios showed a large decrease in rearrangements and elimination of cells. Thus, a possible growth regulation by mechanical force could have a function in releasing the mechanical stress that evolves when all cells have similar growth rates.


Subject(s)
Epithelial Cells/cytology , Mitosis , Stress, Mechanical , Animals , Cell Proliferation , Cell Size , Computer Simulation , Drosophila , Models, Theoretical , Wings, Animal/cytology
10.
Cell Commun Signal ; 11(1): 26, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23590848

ABSTRACT

BACKGROUND: Insulin/insulin-like growth factor signalling (IIS) has been described as one of the major pathways involved in growth control and homeostasis in multicellular organisms. Whereas its core components are well established, less is known about the molecular functions of IIS regulators. The adaptor molecule Lnk/SH2B has been implicated in IIS but the mechanism by which it promotes IIS activity has remained enigmatic. RESULTS: In this study, we analyse genetic and physical interactions among InR, Chico and Lnk in Drosophila tissues. FRET analysis reveals in vivo binding between all three molecules. Genetically, Lnk acts upstream of Chico. We demonstrate that Chico's plasma membrane localisation is ensured by both its PH domain and by the interaction with Lnk. Furthermore, Lnk is able to recruit an intracellular InR fragment to the membrane. CONCLUSIONS: Thus, by acting as a scaffolding molecule that ensures InR and Chico enrichment at the membrane, Lnk provides a fail-safe mechanism for IIS activation.

11.
PLoS Genet ; 6(3): e1000881, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333234

ABSTRACT

Drosophila Lnk is the single ancestral orthologue of a highly conserved family of structurally-related intracellular adaptor proteins, the SH2B proteins. As adaptors, they lack catalytic activity but contain several protein-protein interaction domains, thus playing a critical role in signal transduction from receptor tyrosine kinases to form protein networks. Physiological studies of SH2B function in mammals have produced conflicting data. However, a recent study in Drosophila has shown that Lnk is an important regulator of the insulin/insulin-like growth factor (IGF)-1 signaling (IIS) pathway during growth, functioning in parallel to the insulin receptor substrate, Chico. As this pathway also has an evolutionary conserved role in the determination of organism lifespan, we investigated whether Lnk is required for normal lifespan in Drosophila. Phenotypic analysis of mutants for Lnk revealed that loss of Lnk function results in increased lifespan and improved survival under conditions of oxidative stress and starvation. Starvation resistance was found to be associated with increased metabolic stores of carbohydrates and lipids indicative of impaired metabolism. Biochemical and genetic data suggest that Lnk functions in both the IIS and Ras/Mitogen activated protein Kinase (MapK) signaling pathways. Microarray studies support this model, showing transcriptional feedback onto genes in both pathways as well as indicating global changes in both lipid and carbohydrate metabolism. Finally, our data also suggest that Lnk itself may be a direct target of the IIS responsive transcription factor, dFoxo, and that dFoxo may repress Lnk expression. We therefore describe novel functions for a member of the SH2B protein family and provide the first evidence for potential mechanisms of SH2B regulation. Our findings suggest that IIS signaling in Drosophila may require the activity of a second intracellular adaptor, thereby yielding fundamental new insights into the functioning and role of the IIS pathway in ageing and metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Longevity/physiology , Oxidative Stress , Animals , Body Size , Drosophila melanogaster/genetics , Female , Fertility , Gene Expression Regulation , Insulin/metabolism , MAP Kinase Signaling System/genetics , Male , Mutation/genetics , Oxidative Stress/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Sex Characteristics , Starvation , Transcription, Genetic , ras Proteins/metabolism
12.
PLoS Genet ; 6(5): e1000937, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20463884

ABSTRACT

S6 kinases (S6Ks) act to integrate nutrient and insulin signaling pathways and, as such, function as positive effectors in cell growth and organismal development. However, they also have been shown to play a key role in limiting insulin signaling and in mediating the autophagic response. To identify novel regulators of S6K signaling, we have used a Drosophila-based, sensitized, gain-of-function genetic screen. Unexpectedly, one of the strongest enhancers to emerge from this screen was the nuclear receptor (NR), Drosophila hormone receptor 3 (DHR3), a critical constituent in the coordination of Drosophila metamorphosis. Here we demonstrate that DHR3, through dS6K, also acts to regulate cell-autonomous growth. Moreover, we show that the ligand-binding domain (LBD) of DHR3 is essential for mediating this response. Consistent with these findings, we have identified an endogenous DHR3 isoform that lacks the DBD. These results provide the first molecular link between the dS6K pathway, critical in controlling nutrient-dependent growth, and that of DHR3, a major mediator of ecdysone signaling, which, acting together, coordinate metamorphosis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribosomal Protein S6 Kinases/metabolism , Animals , Drosophila/chemistry , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Female , Gene Expression Regulation, Developmental , Male , Metamorphosis, Biological , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Ribosomal Protein S6 Kinases/genetics , Signal Transduction
13.
CBE Life Sci Educ ; 22(2): ar17, 2023 06.
Article in English | MEDLINE | ID: mdl-36862800

ABSTRACT

Undergraduate biology students' molecular-level understanding of stochastic (also referred to as random or noisy) processes found in biological systems is often limited to those examples discussed in class. Therefore, students frequently display little ability to accurately transfer their knowledge to other contexts. Furthermore, elaborate tools to assess students' understanding of these stochastic processes are missing, despite the fundamental nature of this concept and the increasing evidence demonstrating its importance in biology. Thus, we developed the Molecular Randomness Concept Inventory (MRCI), an instrument composed of nine multiple-choice questions based on students' most prevalent misconceptions, to quantify students' understanding of stochastic processes in biological systems. The MRCI was administered to 67 first-year natural science students in Switzerland. The psychometric properties of the inventory were analyzed using classical test theory and Rasch modeling. Moreover, think-aloud interviews were conducted to ensure response validity. Results indicate that the MRCI yields valid and reliable estimations of students' conceptual understanding of molecular randomness in the higher educational setting studied. Ultimately, the performance analysis sheds light on the extent and the limitations of students' understanding of the concept of stochasticity on a molecular level.


Subject(s)
Knowledge , Students , Humans , Psychometrics
14.
BMC Dev Biol ; 12: 35, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23217079

ABSTRACT

BACKGROUND: The proper balance of autophagy, a lysosome-mediated degradation process, is indispensable for oogenesis in Drosophila. We recently demonstrated that egg development depends on autophagy in the somatic follicle cells (FC), but not in the germline cells (GCs). However, the lack of autophagy only affects oogenesis when FCs are autophagy-deficient but GCs are wild type, indicating that a dysfunctional signaling between soma and germline may be responsible for the oogenesis defects. Thus, autophagy could play an essential role in modulating signal transduction pathways during egg development. RESULTS: Here, we provide further evidence for the necessity of autophagy during oogenesis and demonstrate that autophagy is especially required in subsets of FCs. Generation of autophagy-deficient FCs leads to a wide range of phenotypes that are similar to mutants with defects in the classical cell-cell signaling pathways in the ovary. Interestingly, we observe that loss of autophagy leads to a precocious activation of the Notch pathway in the FCs as monitored by the expression of Cut and Hindsight, two downstream effectors of Notch signaling. CONCLUSION: Our findings point to an unexpected function for autophagy in the modulation of the Notch signaling pathway during Drosophila oogenesis and suggest a function for autophagy in proper receptor activation. Egg development is affected by an imbalance of autophagy between signal sending (germline) and signal receiving cell (FC), thus the lack of autophagy in the germline is likely to decrease the amount of active ligand and accordingly compensates for increased signaling in autophagy-defective follicle cells.


Subject(s)
Autophagy , Drosophila melanogaster/physiology , Oogenesis , Ovarian Follicle/metabolism , Receptors, Notch/metabolism , Animals , Animals, Genetically Modified , Autophagy/genetics , Drosophila Proteins/biosynthesis , Drosophila melanogaster/metabolism , Female , Homeodomain Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Ovarian Follicle/cytology , RNA Interference , RNA, Small Interfering , Signal Transduction , Transcription Factors/biosynthesis
15.
BMC Biotechnol ; 12: 95, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23231964

ABSTRACT

BACKGROUND: Many proteins form insoluble protein aggregates, called "inclusion bodies", when overexpressed in E. coli. This is the biggest obstacle in biotechnology. Ever since the reversible denaturation of proteins by chaotropic agents such as urea or guanidinium hydrochloride had been shown, these compounds were predominantly used to dissolve inclusion bodies. Other denaturants exist but have received much less attention in protein purification. While the anionic, denaturing detergent sodiumdodecylsulphate (SDS) is used extensively in analytical SDS-PAGE, it has rarely been used in preparative purification. RESULTS: Here we present a simple and versatile method to purify insoluble, hexahistidine-tagged proteins under denaturing conditions. It is based on dissolution of overexpressing bacterial cells in a buffer containing sodiumdodecylsulfate (SDS) and whole-lysate denaturation of proteins. The excess of detergent is removed by cooling and centrifugation prior to affinity purification. Host- and overexpressed proteins do not co-precipitate with SDS and the residual concentration of detergent is compatible with affinity purification on Ni/NTA resin. We show that SDS can be replaced with another ionic detergent, Sarkosyl, during purification. Key advantages over denaturing purification in urea or guanidinium are speed, ease of use, low cost of denaturant and the compatibility of buffers with automated FPLC. CONCLUSION: Ionic, denaturing detergents are useful in breaking the solubility barrier, a major obstacle in biotechnology. The method we present yields detergent-denatured protein. Methods to refold proteins from a detergent denatured state are known and therefore we propose that the procedure presented herein will be of general application in biotechnology.


Subject(s)
Detergents/chemistry , Inclusion Bodies/metabolism , Anions/chemistry , Chromatography, Affinity , Escherichia coli/metabolism , Guanidine/chemistry , Histidine/genetics , Histidine/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Denaturation , Protein Refolding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Sodium Dodecyl Sulfate/chemistry , Urea/chemistry
16.
Mol Syst Biol ; 7: 467, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21283143

ABSTRACT

Phosphorylation of the mitogen-activated protein kinase (MAPK) is essential for its enzymatic activity and ability to control multiple substrates inside a cell. According to the current models, control of MAPK phosphorylation is independent of its substrates, which are viewed as mere sensors of MAPK activity. Contrary to this modular view of MAPK signaling, our studies in the Drosophila embryo demonstrate that substrates can regulate the level of MAPK phosphorylation in vivo. We demonstrate that a twofold change in the gene dosage of a single substrate can induce a significant change in the phosphorylation level of MAPK and in the conversion of other substrates. Our results support a model where substrates of MAPK counteract its dephosphorylation by phosphatases. Substrate-dependent control of MAPK phosphorylation is a manifestation of a more general retroactive effect that should be intrinsic to all networks with covalent modification cycles.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila/metabolism , Drosophila/physiology , Drosophila Proteins/metabolism , Dual Specificity Phosphatase 6/metabolism , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , HMGB Proteins/metabolism , Phosphorylation , Repressor Proteins/metabolism , Systems Biology
17.
Mol Syst Biol ; 7: 547, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22068330

ABSTRACT

Genetic analysis in Drosophila melanogaster has been widely used to identify a system of genes that control cell growth in response to insulin and nutrients. Many of these genes encode components of the insulin receptor/target of rapamycin (InR/TOR) pathway. However, the biochemical context of this regulatory system is still poorly characterized in Drosophila. Here, we present the first quantitative study that systematically characterizes the modularity and hormone sensitivity of the interaction proteome underlying growth control by the dInR/TOR pathway. Applying quantitative affinity purification and mass spectrometry, we identified 97 high confidence protein interactions among 58 network components. In all, 22% of the detected interactions were regulated by insulin affecting membrane proximal as well as intracellular signaling complexes. Systematic functional analysis linked a subset of network components to the control of dTORC1 and dTORC2 activity. Furthermore, our data suggest the presence of three distinct dTOR kinase complexes, including the evolutionary conserved dTTT complex (Drosophila TOR, TELO2, TTI1). Subsequent genetic studies in flies suggest a role for dTTT in controlling cell growth via a dTORC1- and dTORC2-dependent mechanism.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Protein Kinases/metabolism , Proteome/metabolism , Receptor, Insulin/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Mass Spectrometry , Protein Interaction Maps , Protein Kinases/genetics , Proteome/genetics , Receptor, Insulin/genetics , Signal Transduction , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
18.
PLoS Biol ; 7(11): e1000236, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19885390

ABSTRACT

Protein modifications play a major role for most biological processes in living organisms. Amino-terminal acetylation of proteins is a common modification found throughout the tree of life: the N-terminus of a nascent polypeptide chain becomes co-translationally acetylated, often after the removal of the initiating methionine residue. While the enzymes and protein complexes involved in these processes have been extensively studied, only little is known about the biological function of such N-terminal modification events. To identify common principles of N-terminal acetylation, we analyzed the amino-terminal peptides from proteins extracted from Drosophila Kc167 cells. We detected more than 1,200 mature protein N-termini and could show that N-terminal acetylation occurs in insects with a similar frequency as in humans. As the sole true determinant for N-terminal acetylation we could extract the (X)PX rule that indicates the prevention of acetylation under all circumstances. We could show that this rule can be used to genetically engineer a protein to study the biological relevance of the presence or absence of an acetyl group, thereby generating a generic assay to probe the functional importance of N-terminal acetylation. We applied the assay by expressing mutated proteins as transgenes in cell lines and in flies. Here, we present a straightforward strategy to systematically study the functional relevance of N-terminal acetylations in cells and whole organisms. Since the (X)PX rule seems to be of general validity in lower as well as higher eukaryotes, we propose that it can be used to study the function of N-terminal acetylation in all species.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Protein Processing, Post-Translational/physiology , Acetylation , Alanine/genetics , Alanine/metabolism , Animals , Animals, Genetically Modified , Binding Sites/genetics , Blotting, Western , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Cell Line , Databases, Protein , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , HeLa Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mutation , Protein Biosynthesis , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , Transgenes/genetics
19.
PLoS Genet ; 5(8): e1000596, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19680438

ABSTRACT

Insulin/insulin-like growth factor signaling (IIS) plays a pivotal role in the regulation of growth at the cellular and the organismal level during animal development. Flies with impaired IIS are developmentally delayed and small due to fewer and smaller cells. In the search for new growth-promoting genes, we identified mutations in the gene encoding Lnk, the single fly member of the SH2B family of adaptor molecules. Flies lacking lnk function are viable but severely reduced in size. Furthermore, lnk mutants display phenotypes reminiscent of reduced IIS, such as developmental delay, female sterility, and accumulation of lipids. Genetic epistasis analysis places lnk downstream of the insulin receptor (InR) and upstream of phosphoinositide 3-kinase (PI3K) in the IIS cascade, at the same level as chico (encoding the single fly insulin receptor substrate [IRS] homolog). Both chico and lnk mutant larvae display a similar reduction in IIS activity as judged by the localization of a PIP(3) reporter and the phosphorylation of protein kinase B (PKB). Furthermore, chico; lnk double mutants are synthetically lethal, suggesting that Chico and Lnk fulfill independent but partially redundant functions in the activation of PI3K upon InR stimulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Multigene Family , Signal Transduction , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Body Size , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Insulin/genetics , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Mutation , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sequence Alignment , src Homology Domains
20.
Nat Cell Biol ; 5(6): 559-65, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766775

ABSTRACT

Understanding the mechanisms through which multicellular organisms regulate cell, organ and body growth is of relevance to developmental biology and to research on growth-related diseases such as cancer. Here we describe a new effector in growth control, the small GTPase Rheb (Ras homologue enriched in brain). Mutations in the Drosophila melanogaster Rheb gene were isolated as growth-inhibitors, whereas overexpression of Rheb promoted cell growth. Our genetic and biochemical analyses suggest that Rheb functions downstream of the tumour suppressors Tsc1 (tuberous sclerosis 1)-Tsc2 in the TOR (target of rapamycin) signalling pathway to control growth, and that a major effector of Rheb function is ribosomal S6 kinase (S6K).


Subject(s)
Cell Division/genetics , Drosophila Proteins/metabolism , Growth Substances/metabolism , Monomeric GTP-Binding Proteins/physiology , Neuropeptides/physiology , Ribosomal Protein S6 Kinases/metabolism , Animals , Cell Division/physiology , Cell Size/genetics , Cell Size/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye/ultrastructure , Female , Gene Deletion , Genes, Insect , Genes, Tumor Suppressor , Growth Substances/genetics , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Ras Homolog Enriched in Brain Protein , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases/genetics , Signal Transduction , Transcriptional Activation , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL