Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Bio Protoc ; 12(13)2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35937934

ABSTRACT

Understanding protein-protein interactions (PPIs) and interactome networks is essential to reveal molecular mechanisms mediating various cellular processes. The most common method to study PPIs in vivo is affinity purification combined with mass spectrometry (AP-MS). Although AP-MS is a powerful method, loss of weak and transient interactions is still a major limitation. Proximity labeling (PL) techniques have been developed as alternatives to overcome these limitations. Proximity-dependent biotin identification (BioID) is one such widely used PL method. The first-generation BioID enzyme BirA*, a promiscuous bacterial biotin ligase, has been effectively used in cultured mammalian cells; however, relatively slow enzyme kinetics make it less effective for temporal analysis of protein interactions. In addition, BirA* exhibits reduced activity at temperatures below 37°C, further restricting its use in intact organisms cultured at lower optimal growth temperatures ( e.g., Drosophila melanogaster ). TurboID, miniTurbo, and BirA*-G3 are next generation BirA* variants with improved catalytic activity, allowing investigators to use this powerful tool in model systems such as flies. Here, we describe a detailed experimental workflow to efficiently identify the proximal proteome (proximitome) of a protein of interest (POI) in the Drosophila brain using CRISPR/Cas9-induced homology-directed repair (HDR) strategies to endogenously tag the POI with next generation BioID enzymes.

2.
Dis Model Mech ; 15(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35972154

ABSTRACT

Activating anaplastic lymphoma kinase (ALK) receptor tyrosine kinase (RTK) mutations occur in pediatric neuroblastoma and are associated with poor prognosis. To study ALK-activating mutations in a genetically controllable system, we employed CRIPSR/Cas9, incorporating orthologs of the human oncogenic mutations ALKF1174L and ALKY1278S in the Drosophila Alk locus. AlkF1251L and AlkY1355S mutant Drosophila exhibited enhanced Alk signaling phenotypes, but unexpectedly depended on the Jelly belly (Jeb) ligand for activation. Both AlkF1251L and AlkY1355S mutant larval brains displayed hyperplasia, represented by increased numbers of Alk-positive neurons. Despite this hyperplasic phenotype, no brain tumors were observed in mutant animals. We showed that hyperplasia in Alk mutants was not caused by significantly increased rates of proliferation, but rather by decreased levels of apoptosis in the larval brain. Using single-cell RNA sequencing, we identified perturbations during temporal fate specification in AlkY1355S mutant mushroom body lineages. These findings shed light on the role of Alk in neurodevelopmental processes and highlight the potential of Alk-activating mutations to perturb specification and promote survival in neuronal lineages. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Anaplastic Lymphoma Kinase , Cell Differentiation , Drosophila Proteins , Neurons , Anaplastic Lymphoma Kinase/genetics , Animals , Child , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Humans , Hyperplasia , Mutation , Neurons/cytology , Receptor Protein-Tyrosine Kinases/genetics
3.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34905617

ABSTRACT

Development of the Drosophila visceral muscle depends on Anaplastic Lymphoma Kinase (Alk) receptor tyrosine kinase (RTK) signaling, which specifies founder cells (FCs) in the circular visceral mesoderm (VM). Although Alk activation by its ligand Jelly Belly (Jeb) is well characterized, few target molecules have been identified. Here, we used targeted DamID (TaDa) to identify Alk targets in embryos overexpressing Jeb versus embryos with abrogated Alk activity, revealing differentially expressed genes, including the Snail/Scratch family transcription factor Kahuli (Kah). We confirmed Kah mRNA and protein expression in the VM, and identified midgut constriction defects in Kah mutants similar to those of pointed (pnt). ChIP and RNA-Seq data analysis defined a Kah target-binding site similar to that of Snail, and identified a set of common target genes putatively regulated by Kah and Pnt during midgut constriction. Taken together, we report a rich dataset of Alk-responsive loci in the embryonic VM and functionally characterize the role of Kah in the regulation of embryonic midgut morphogenesis.


Subject(s)
Anaplastic Lymphoma Kinase , DNA-Binding Proteins , Drosophila Proteins , Embryonic Development , Nerve Tissue Proteins , Proto-Oncogene Proteins , Transcription Factors , Animals , Anaplastic Lymphoma Kinase/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila Proteins/genetics , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Mesoderm/growth & development , Mesoderm/metabolism , Muscle Development/genetics , Muscles/metabolism , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Transcription Factors/genetics
4.
J Mol Biol ; 433(23): 167282, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34624297

ABSTRACT

Anaplastic lymphoma kinase (Alk) is an evolutionary conserved receptor tyrosine kinase belonging to the insulin receptor superfamily. In addition to its well-studied role in cancer, numerous studies have revealed that Alk signaling is associated with a variety of complex traits such as: regulation of growth and metabolism, hibernation, regulation of neurotransmitters, synaptic coupling, axon targeting, decision making, memory formation and learning, alcohol use disorder, as well as steroid hormone metabolism. In this study, we used BioID-based in vivo proximity labeling to identify molecules that interact with Alk in the Drosophila central nervous system (CNS). To do this, we used CRISPR/Cas9 induced homology-directed repair (HDR) to modify the endogenous Alk locus to produce first and next generation Alk::BioID chimeras. This approach allowed identification of Alk proximitomes under physiological conditions and without overexpression. Our results show that the next generation of BioID proteins (TurboID and miniTurbo) outperform the first generation BirA* fusion in terms of labeling speed and efficiency. LC-MS3-based BioID screening of AlkTurboID and AlkminiTurbo larval brains revealed an extensive neuronal Alk proximitome identifying numerous potential components of Alk signaling complexes. Validation of Alk proximitome candidates further revealed co-expression of Stardust (Sdt), Discs large 1 (Dlg1), Syntaxin (Syx) and Rugose (Rg) with Alk in the CNS and identified the protein-tyrosine-phosphatase Corkscrew (Csw) as a modulator of Alk signaling.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Brain/embryology , Brain/metabolism , Organogenesis , Proteome , Proteomics , Anaplastic Lymphoma Kinase/genetics , Animals , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Gene Expression Regulation, Developmental , Organogenesis/genetics , Proteomics/methods , Signal Transduction
5.
J Mol Biol ; 433(19): 167158, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34273398

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that is mutated in approximately 10% of pediatric neuroblastoma (NB). To shed light on ALK-driven signaling processes, we employed BioID-based in vivo proximity labeling to identify molecules that interact intracellularly with ALK. NB-derived SK-N-AS and SK-N-BE(2) cells expressing inducible ALK-BirA* fusion proteins were generated and stimulated with ALKAL ligands in the presence and absence of the ALK tyrosine kinase inhibitor (TKI) lorlatinib. LC/MS-MS analysis identified multiple proteins, including PEAK1 and SHP2, which were validated as ALK interactors in NB cells. Further analysis of the ALK-SHP2 interaction confirmed that the ALK-SHP2 interaction as well as SHP2-Y542 phosphorylation was dependent on ALK activation. Use of the SHP2 inhibitors, SHP099 and RMC-4550, resulted in inhibition of cell growth in ALK-driven NB cells. In addition, we noted a strong synergistic effect of combined ALK and SHP2 inhibition that was specific to ALK-driven NB cells, suggesting a potential therapeutic option for ALK-driven NB.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Neuroblastoma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein-Tyrosine Kinases/metabolism , Proteomics/methods , Aminopyridines/pharmacology , Animals , Cell Line, Tumor , Chromatography, Liquid , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Lactams/pharmacology , PC12 Cells , Phosphorylation , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Tandem Mass Spectrometry
6.
Sci Rep ; 10(1): 14954, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917927

ABSTRACT

Anaplastic lymphoma kinase (Alk) is a receptor tyrosine kinase of the insulin receptor super-family that functions as oncogenic driver in a range of human cancers such as neuroblastoma. In order to investigate mechanisms underlying Alk oncogenic signaling, we conducted a genetic suppressor screen in Drosophila melanogaster. Our screen identified multiple loci important for Alk signaling, including members of Ras/Raf/ERK-, Pi3K-, and STAT-pathways as well as tailless (tll) and foxo whose orthologues NR2E1/TLX and FOXO3 are transcription factors implicated in human neuroblastoma. Many of the identified suppressors were also able to modulate signaling output from activated oncogenic variants of human ALK, suggesting that our screen identified targets likely relevant in a wide range of contexts. Interestingly, two misexpression alleles of wallenda (wnd, encoding a leucine zipper bearing kinase similar to human DLK and LZK) were among the strongest suppressors. We show that Alk expression leads to a growth advantage and induces cell death in surrounding cells. Our results suggest that Alk activity conveys a competitive advantage to cells, which can be reversed by over-expression of the JNK kinase kinase Wnd.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Drosophila Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Signal Transduction , Anaplastic Lymphoma Kinase/genetics , Animals , Cell Death , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , MAP Kinase Kinase Kinases/genetics
7.
Development ; 146(2)2019 01 16.
Article in English | MEDLINE | ID: mdl-30567930

ABSTRACT

Basement membranes (BMs) are specialized layers of extracellular matrix (ECM) mainly composed of Laminin, type IV Collagen, Perlecan and Nidogen/entactin (NDG). Recent in vivo studies challenged the initially proposed role of NDG as a major ECM linker molecule by revealing dispensability for viability and BM formation. Here, we report the characterization of the single Ndg gene in Drosophila. Embryonic Ndg expression was primarily observed in mesodermal tissues and the chordotonal organs, whereas NDG protein localized to all BMs. Although loss of Laminin strongly affected BM localization of NDG, Ndg-null mutants exhibited no overt changes in the distribution of BM components. Although Drosophila Ndg mutants were viable, loss of NDG led to ultrastructural BM defects that compromised barrier function and stability in vivo Moreover, loss of NDG impaired larval crawling behavior and reduced responses to vibrational stimuli. Further morphological analysis revealed accompanying defects in the larval peripheral nervous system, especially in the chordotonal organs and the neuromuscular junction (NMJ). Taken together, our analysis suggests that NDG is not essential for BM assembly but mediates BM stability and ECM-dependent neural plasticity during Drosophila development.


Subject(s)
Basement Membrane/metabolism , Body Patterning , Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Extracellular Matrix Proteins/metabolism , Nervous System/embryology , Nervous System/metabolism , Animals , Basement Membrane/ultrastructure , Behavior, Animal , Biomechanical Phenomena , Calcium-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Extracellular Matrix Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Laminin/metabolism , Larva/genetics , Neuromuscular Junction/pathology , Peripheral Nervous System/embryology , Peripheral Nervous System/pathology , Permeability , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vibration
8.
Sci Signal ; 10(502)2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29066538

ABSTRACT

In Drosophila melanogaster, the receptor tyrosine kinase (RTK) anaplastic lymphoma kinase (Alk) and its ligand jelly belly (Jeb) are required to specify muscle founder cells in the visceral mesoderm. We identified a critical role for the scaffolding protein Cnk (connector enhancer of kinase suppressor of Ras) in this signaling pathway. Embryos that ectopically expressed the minimal Alk interaction region in the carboxyl terminus of Cnk or lacked maternal and zygotic cnk did not generate visceral founder cells or a functional gut musculature, phenotypes that resemble those of jeb and Alk mutants. Deletion of the entire Alk-interacting region in the cnk locus affected the Alk signaling pathway in the visceral mesoderm and not other RTK signaling pathways in other tissues. In addition, the Cnk-interacting protein Aveugle (Ave) was critical for Alk signaling in the developing visceral mesoderm. Alk signaling stimulates the MAPK/ERK pathway, but the scaffolding protein Ksr, which facilitates activation of this pathway, was not required to promote visceral founder cell specification. Thus, Cnk and Ave represent critical molecules downstream of Alk, and their loss genocopies the lack of visceral founder cell specification of Alk and jeb mutants, indicating their essential roles in Alk signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Receptor Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Anaplastic Lymphoma Kinase , Animals , Databases, Nucleic Acid , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Humans , Mesoderm/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Sequence Deletion , Zygote
9.
PLoS Genet ; 13(4): e1006617, 2017 04.
Article in English | MEDLINE | ID: mdl-28369060

ABSTRACT

The Anaplastic Lymphoma Kinase (Alk) receptor tyrosine kinase (RTK) plays a critical role in the specification of founder cells (FCs) in the Drosophila visceral mesoderm (VM) during embryogenesis. Reporter gene and CRISPR/Cas9 deletion analysis reveals enhancer regions in and upstream of the Alk locus that influence tissue-specific expression in the amnioserosa (AS), the VM and the epidermis. By performing high throughput yeast one-hybrid screens (Y1H) with a library of Drosophila transcription factors (TFs) we identify Odd-paired (Opa), the Drosophila homologue of the vertebrate Zic family of TFs, as a novel regulator of embryonic Alk expression. Further characterization identifies evolutionarily conserved Opa-binding cis-regulatory motifs in one of the Alk associated enhancer elements. Employing Alk reporter lines as well as CRISPR/Cas9-mediated removal of regulatory elements in the Alk locus, we show modulation of Alk expression by Opa in the embryonic AS, epidermis and VM. In addition, we identify enhancer elements that integrate input from additional TFs, such as Binou (Bin) and Bagpipe (Bap), to regulate VM expression of Alk in a combinatorial manner. Taken together, our data show that the Opa zinc finger TF is a novel regulator of embryonic Alk expression.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Anaplastic Lymphoma Kinase , Animals , Animals, Genetically Modified , Binding Sites , CRISPR-Cas Systems , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism
10.
Oncotarget ; 8(7): 11566-11578, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28030793

ABSTRACT

Activating mutations in full length anaplastic lymphoma kinase (ALK) have been reported in neuroblastoma and in anaplastic thyroid cancer. ALK-L1198F and ALK-G1201E mutations were originally identified in anaplastic thyroid cancer (ATC) and characterized as constitutively activating mutations. In this study, we employed in vitro cell culture assays together with biochemical and in vivo Drosophila analyses to characterize their sensitivity to either activation by the FAM150A (AUG-ß) and FAM150B (AUG-α) ALK ligands or inhibition by ALK inhibitors. Here we report that neither ALK-L1198F nor ALK-G1201E mutations result in ligand independent gain-of-function (GOF) activity in either in vitro biochemical analysis or the various model systems employed. ALK-L1198F is activated by the FAM150 (AUG) ligands and its ligand-dependant activity is similar to the wild type full length ALK receptor. ALK-G1201E is only very weakly activated by the FAM150 (AUG) ligands, most likely due to impaired protein stability. We conclude that neither ALK-L1198F nor ALK-G1201E displays ligand independent kinase activity, with ALK-L1198F belonging to the class of ligand dependent ALK mutations which are not constitutively active but that responds to ligand activation, while the ALK-G1201E mutation generates an unstable receptor with very low levels of kinase activity.


Subject(s)
Drosophila/genetics , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Anaplastic Lymphoma Kinase , Animals , Humans , Ligands , Thyroid Carcinoma, Anaplastic/enzymology , Thyroid Neoplasms/enzymology
11.
Elife ; 4: e09811, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26418745

ABSTRACT

Aberrant activation of anaplastic lymphoma kinase (ALK) has been described in a range of human cancers, including non-small cell lung cancer and neuroblastoma (Hallberg and Palmer, 2013). Vertebrate ALK has been considered to be an orphan receptor and the identity of the ALK ligand(s) is a critical issue. Here we show that FAM150A and FAM150B are potent ligands for human ALK that bind to the extracellular domain of ALK and in addition to activation of wild-type ALK are able to drive 'superactivation' of activated ALK mutants from neuroblastoma. In conclusion, our data show that ALK is robustly activated by the FAM150A/B ligands and provide an opportunity to develop ALK-targeted therapies in situations where ALK is overexpressed/activated or mutated in the context of the full length receptor.


Subject(s)
Cytokines/metabolism , Enzyme Activation , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Cell Line , Humans , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA
12.
BMC Cell Biol ; 15: 27, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25000973

ABSTRACT

BACKGROUND: The visceral musculature of Drosophila larvae comprises circular visceral muscles tightly interwoven with longitudinal visceral muscles. During myogenesis, the circular muscles arise by one-to-one fusion of a circular visceral founder cell (FC) with a visceral fusion-competent myoblast (FCM) from the trunk visceral mesoderm, and longitudinal muscles arise from FCs of the caudal visceral mesoderm. Longitudinal FCs migrate anteriorly under guidance of fibroblast growth factors during embryogenesis; it is proposed that they fuse with FCMs from the trunk visceral mesoderm to give rise to syncytia containing up to six nuclei. RESULTS: Using fluorescence in situ hybridization and immunochemical analyses, we investigated whether these fusion events during migration use the same molecular repertoire and cellular components as fusion-restricted myogenic adhesive structure (FuRMAS), the adhesive signaling center that mediates myoblast fusion in the somatic mesoderm. Longitudinal muscles were formed by the fusion of one FC with Sns-positive FCMs, and defects in FCM specification led to defects in longitudinal muscle formation. At the fusion sites, Duf/Kirre and the adaptor protein Rols7 accumulated in longitudinal FCs, and Blow and F-actin accumulated in FCMs. The accumulation of these four proteins at the fusion sites argues for FuRMAS-like adhesion and signaling centers. Longitudinal fusion was disturbed in rols and blow single, and scar wip double mutants. Mutants of wasp or its interaction partner wip had no defects in longitudinal fusion. CONCLUSIONS: Our results indicated that all embryonic fusion events depend on the same cell-adhesion molecules, but that the need for Rols7 and regulators of F-actin distinctly differs. Rols7 was required for longitudinal visceral and somatic myoblast fusion but not for circular visceral fusion. Importantly, longitudinal fusion depended on Kette and SCAR/Wave but was independent of WASp-dependent Arp2/3 activation. Thus, the complexity of the players involved in muscle formation increases from binucleated circular muscles to longitudinal visceral muscles to somatic muscles.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Myoblasts/cytology , Animals , Animals, Genetically Modified , Cell Movement , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , Muscle Development , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/embryology , Muscles/metabolism , Myoblasts/metabolism
13.
PLoS One ; 9(5): e96702, 2014.
Article in English | MEDLINE | ID: mdl-24792484

ABSTRACT

In this study, we aimed to identify molecular mechanisms involved in the specification of the 4d (mesentoblast) lineage in Platynereis dumerilii. We employ RT-PCR and in situ hybridization against the Platynereis dumerilii twist homolog (Pdu-twist) to reveal mesodermal specification within this lineage. We show that Pdu-twist mRNA is already maternally distributed. After fertilization, ooplasmatic segregation leads to relocation of Pdu-twist transcripts into the somatoblast (2d) lineage and 4d, indicating that the maternal component of Pdu-twist might be an important prerequisite for further mesoderm specification but does not represent a defining characteristic of the mesentoblast. However, after the primordial germ cells have separated from the 4d lineage, zygotic transcription of Pdu-twist is exclusively observed in the myogenic progenitors, suggesting that mesodermal specification occurs after the 4d stage. Previous studies on spiral cleaving embryos revealed a spatio-temporal correlation between the 4d lineage and the activity of an embryonic organizer that is capable to induce the developmental fates of certain micromeres. This has raised the question if specification of the 4d lineage could be connected to the organizer activity. Therefore, we aimed to reveal the existence of such a proposed conserved organizer in Platynereis employing antibody staining against dpERK. In contrast to former observations in other spiralian embryos, activation of MAPK signaling during 2d and 4d formation cannot be detected which questions the existence of a conserved connection between organizer function and specification of the 4d lineage. However, our experiments unveil robust MAPK activation in the prospective nephroblasts as well as in the macromeres and some micromeres at the blastopore in gastrulating embryos. Inhibition of MAPK activation leads to larvae with a shortened body axis, defects in trunk muscle spreading and improper nervous system condensation, indicating a critical function for MAPK signaling for the reorganization of embryonic tissues during the gastrulation process.


Subject(s)
Embryo, Nonmammalian/embryology , Enzyme Activation , Gene Expression Regulation, Developmental , Mitogen-Activated Protein Kinases/metabolism , Polychaeta/embryology , RNA, Messenger/genetics , Twist-Related Protein 1/genetics , Animals , Embryo, Nonmammalian/metabolism , Female , Polychaeta/genetics , RNA, Messenger/analysis
14.
Development ; 140(15): 3156-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824577

ABSTRACT

The Jelly belly (Jeb)/Anaplastic Lymphoma Kinase (Alk) signalling pathway regulates myoblast fusion in the circular visceral mesoderm (VM) of Drosophila embryos via specification of founder cells. However, only a limited number of target molecules for this pathway are described. We have investigated the role of the Lame Duck (Lmd) transcription factor in VM development in relationship to Jeb/Alk signal transduction. We show that Alk signalling negatively regulates Lmd activity post-transcriptionally through the MEK/MAPK (ERK) cascade resulting in a relocalisation of Lmd protein from the nucleus to cytoplasm. It has previously been shown that downregulation of Lmd protein is necessary for the correct specification of founder cells. In the visceral mesoderm of lmd mutant embryos, fusion-competent myoblasts seem to be converted to 'founder-like' cells that are still able to build a gut musculature even in the absence of fusion. The ability of Alk signalling to downregulate Lmd protein requires the N-terminal 140 amino acids, as a Lmd(141-866) mutant remains nuclear in the presence of active ALK and is able to drive robust expression of the Lmd downstream target Vrp1 in the developing VM. Our results suggest that Lmd is a target of Jeb/Alk signalling in the VM of Drosophila embryos.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Myogenic Regulatory Factors/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Active Transport, Cell Nucleus , Anaplastic Lymphoma Kinase , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Carrier Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Insect , MAP Kinase Signaling System , Mesoderm/embryology , Mesoderm/metabolism , Models, Biological , Muscle Development , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myogenic Regulatory Factors/chemistry , Myogenic Regulatory Factors/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction
15.
Dev Genes Evol ; 223(5): 319-28, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23817621

ABSTRACT

The basic helix-loop-helix transcription factor twist plays a key role during mesoderm development in Bilateria. In this study, we identified a twist ortholog in the polychaete annelid Platynereis dumerilii and analyze its expression during larval development, postlarval growth up to the adult stage, and caudal regeneration after amputation of posterior segments. At late larval stages, Pdu-twist is expressed in the mesodermal anlagen and in developing muscles. During adulthood and caudal regeneration, Pdu-twist is expressed in the posterior growth zone, in mesodermal cells within the newly forming segments and budding parapodia. Our results indicate that Pdu-twist is involved in mesoderm formation during larval development, posterior growth, and caudal regeneration.


Subject(s)
Polychaeta/embryology , Polychaeta/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Molecular Sequence Data , Phylogeny , Twist-Related Protein 1/chemistry
16.
Cell Mol Life Sci ; 69(2): 267-82, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21387145

ABSTRACT

In Drosophila, four genes encode for laminin subunits and the formation of two laminin heterotrimers has been postulated. We report the identification of mutations in the Drosophila LamininB2 (LanB2) gene that encodes for the only laminin γ subunit and is found in both heterotrimers. We describe their effects on embryogenesis, in particular the differentiation of visceral tissues with respect to the ECM. Analysis of mesoderm endoderm interaction indicates disrupted basement membranes and defective endoderm migration, which finally interferes with visceral myotube stretching. Extracellular deposition of laminin is blocked due to the loss of the LanB2 subunit, resulting in an abnormal distribution of ECM components. Our data, concerning the different function of both trimers during organogenesis, suggest that these trimers might act in a cumulative way and probably at multiple steps during ECM assembly. We also observed genetic interactions with kon-tiki and thrombospondin, indicating a role for laminin during muscle attachment.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Laminin/metabolism , Mesoderm/embryology , Organogenesis , Animals , Basement Membrane/embryology , Cell Differentiation , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Laminin/genetics , Mesoderm/metabolism , Mutation , Thrombospondins/metabolism
17.
Mech Dev ; 126(8-9): 721-36, 2009.
Article in English | MEDLINE | ID: mdl-19463947

ABSTRACT

In this study we describe the morphological and genetic analysis of the Drosophila mutant gürtelchen (gurt). gurt was identified by screening an EMS collection for novel mutations affecting visceral mesoderm development and was named after the distinct belt shaped visceral phenotype. Interestingly, determination of visceral cell identities and subsequent visceral myoblast fusion is not affected in mutant embryos indicating a later defect in visceral development. gurt is in fact a new huckebein (hkb) allele and as such exhibits nearly complete loss of endodermal derived structures. Targeted ablation of the endodermal primordia produces a phenotype that resembles the visceral defects observed in huckebein(gürtelchen) (hkb(gurt)) mutant embryos. It was shown previously that visceral mesoderm development requires complex interactions between visceral myoblasts and adjacent tissues. Signals from the neighbouring somatic myoblasts play an important role in cell type determination and are a prerequisite for visceral muscle fusion. Furthermore, the visceral mesoderm is known to influence endodermal migration and midgut epithelium formation. Our analyses of the visceral phenotype of hkb(gurt) mutant embryos reveal that the adjacent endoderm plays a critical role in the later stages of visceral muscle development, and is required for visceral muscle elongation and outgrowth after proper myoblast fusion.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Endoderm/metabolism , Mesoderm/metabolism , Muscles/embryology , Muscles/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Genes, Insect , Microscopy, Fluorescence/methods , Models, Biological , Models, Genetic , Mutation , Myoblasts/cytology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...