Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep ; 39(1): 110618, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385751

ABSTRACT

Neurons in the developing brain express many different cell adhesion molecules (CAMs) on their surfaces. CAM-binding affinities can vary by more than 200-fold, but the significance of these variations is unknown. Interactions between the immunoglobulin superfamily CAM DIP-α and its binding partners, Dpr10 and Dpr6, control synaptic targeting and survival of Drosophila optic lobe neurons. We design mutations that systematically change interaction affinity and analyze function in vivo. Reducing affinity causes loss-of-function phenotypes whose severity scales with the magnitude of the change. Synaptic targeting is more sensitive to affinity reduction than is cell survival. Increasing affinity rescues neurons that would normally be culled by apoptosis. By manipulating CAM expression together with affinity, we show that the key parameter controlling circuit assembly is surface avidity, which is the strength of adherence between cell surfaces. We conclude that CAM binding affinities and expression levels are finely tuned for function during development.


Subject(s)
Drosophila Proteins , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Survival , Drosophila/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Neurons/metabolism
2.
Elife ; 82019 02 04.
Article in English | MEDLINE | ID: mdl-30714901

ABSTRACT

For animals to perform coordinated movements requires the precise organization of neural circuits controlling motor function. Motor neurons (MNs), key components of these circuits, project their axons from the central nervous system and form precise terminal branching patterns at specific muscles. Focusing on the Drosophila leg neuromuscular system, we show that the stereotyped terminal branching of a subset of MNs is mediated by interacting transmembrane Ig superfamily proteins DIP-α and Dpr10, present in MNs and target muscles, respectively. The DIP-α/Dpr10 interaction is needed only after MN axons reach the vicinity of their muscle targets. Live imaging suggests that precise terminal branching patterns are gradually established by DIP-α/Dpr10-dependent interactions between fine axon filopodia and developing muscles. Further, different leg MNs depend on the DIP-α and Dpr10 interaction to varying degrees that correlate with the morphological complexity of the MNs and their muscle targets.


Subject(s)
Drosophila Proteins/genetics , Motor Neurons/physiology , Neurogenesis/genetics , Transcription Factors/genetics , Animals , Axons/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Motor Neurons/metabolism , Neurons, Efferent/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Transcription Factors/metabolism
3.
Elife ; 82019 02 04.
Article in English | MEDLINE | ID: mdl-30714906

ABSTRACT

The Drosophila larval neuromuscular system provides an ideal context in which to study synaptic partner choice, because it contains a small number of pre- and postsynaptic cells connected in an invariant pattern. The discovery of interactions between two subfamilies of IgSF cell surface proteins, the Dprs and the DIPs, provided new candidates for cellular labels controlling synaptic specificity. Here we show that DIP-α is expressed by two identified motor neurons, while its binding partner Dpr10 is expressed by postsynaptic muscle targets. Removal of either DIP-α or Dpr10 results in loss of specific axonal branches and NMJs formed by one motor neuron, MNISN-1s, while other branches of the MNISN-1s axon develop normally. The temporal and spatial expression pattern of dpr10 correlates with muscle innervation by MNISN-1s during embryonic development. We propose a model whereby DIP-α and Dpr10 on opposing synaptic partners interact with each other to generate proper motor neuron connectivity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intercellular Signaling Peptides and Proteins/genetics , Motor Neurons/physiology , Transcription Factors/genetics , Animals , Axons/metabolism , Axons/physiology , Drosophila melanogaster/physiology , Larva/genetics , Larva/growth & development , Membrane Proteins/genetics , Membrane Proteins/physiology , Muscles/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuronal Plasticity/genetics , Neuropeptides/genetics
4.
Neuron ; 100(6): 1369-1384.e6, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30467079

ABSTRACT

Drosophila Dpr (21 paralogs) and DIP proteins (11 paralogs) are cell recognition molecules of the immunoglobulin superfamily (IgSF) that form a complex protein interaction network. DIP and Dpr proteins are expressed in a synaptic layer-specific fashion in the visual system. How interactions between these proteins regulate layer-specific synaptic circuitry is not known. Here we establish that DIP-α and its interacting partners Dpr6 and Dpr10 regulate multiple processes, including arborization within layers, synapse number, layer specificity, and cell survival. We demonstrate that heterophilic binding between Dpr6/10 and DIP-α and homophilic binding between DIP-α proteins promote interactions between processes in vivo. Knockin mutants disrupting the DIP/Dpr binding interface reveal a role for these proteins during normal development, while ectopic expression studies support an instructive role for interactions between DIPs and Dprs in circuit development. These studies support an important role for the DIP/Dpr protein interaction network in regulating cell-type-specific connectivity patterns.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Neuropil/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Drosophila , Drosophila Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Medulla Oblongata/cytology , Medulla Oblongata/growth & development , Mutation/genetics , Protein Interaction Maps , Surface Plasmon Resonance , Transcription Factors/genetics , Transfection , Visual Pathways/metabolism
5.
Neuron ; 100(6): 1385-1400.e6, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30467080

ABSTRACT

Binding between DIP and Dpr neuronal recognition proteins has been proposed to regulate synaptic connections between lamina and medulla neurons in the Drosophila visual system. Each lamina neuron was previously shown to express many Dprs. Here, we demonstrate, by contrast, that their synaptic partners typically express one or two DIPs, with binding specificities matched to the lamina neuron-expressed Dprs. A deeper understanding of the molecular logic of DIP/Dpr interaction requires quantitative studies on the properties of these proteins. We thus generated a quantitative affinity-based DIP/Dpr interactome for all DIP/Dpr protein family members. This revealed a broad range of affinities and identified homophilic binding for some DIPs and some Dprs. These data, along with full-length ectodomain DIP/Dpr and DIP/DIP crystal structures, led to the identification of molecular determinants of DIP/Dpr specificity. This structural knowledge, along with a comprehensive set of quantitative binding affinities, provides new tools for functional studies in vivo.


Subject(s)
Drosophila Proteins/metabolism , Medulla Oblongata/cytology , Neurons/metabolism , Visual Pathways/cytology , Animals , Animals, Genetically Modified , Cell Communication , Drosophila Proteins/genetics , Drosophila melanogaster , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Protein Binding , Surface Plasmon Resonance , Transfection
6.
Neuron ; 89(3): 480-93, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26844831

ABSTRACT

Cell recognition molecules are key regulators of neural circuit assembly. The Dscam family of recognition molecules in Drosophila has been shown to regulate interactions between neurons through homophilic repulsion. This is exemplified by Dscam1 and Dscam2, which together repel dendrites of lamina neurons, L1 and L2, in the visual system. By contrast, here we show that Dscam2 directs dendritic targeting of another lamina neuron, L4, through homophilic adhesion. Through live imaging and genetic mosaics to dissect interactions between specific cells, we show that Dscam2 is required in L4 and its target cells for correct dendritic targeting. In a genetic screen, we identified Dscam4 as another regulator of L4 targeting which acts with Dscam2 in the same pathway to regulate this process. This ensures tiling of the lamina neuropil through heterotypic interactions. Thus, different combinations of Dscam proteins act through distinct mechanisms in closely related neurons to pattern neural circuits.


Subject(s)
Dendrites/physiology , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Neural Cell Adhesion Molecules/physiology , Alleles , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Mosaicism , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics
7.
Cell ; 163(7): 1756-69, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26687360

ABSTRACT

Information processing relies on precise patterns of synapses between neurons. The cellular recognition mechanisms regulating this specificity are poorly understood. In the medulla of the Drosophila visual system, different neurons form synaptic connections in different layers. Here, we sought to identify candidate cell recognition molecules underlying this specificity. Using RNA sequencing (RNA-seq), we show that neurons with different synaptic specificities express unique combinations of mRNAs encoding hundreds of cell surface and secreted proteins. Using RNA-seq and protein tagging, we demonstrate that 21 paralogs of the Dpr family, a subclass of immunoglobulin (Ig)-domain containing proteins, are expressed in unique combinations in homologous neurons with different layer-specific synaptic connections. Dpr interacting proteins (DIPs), comprising nine paralogs of another subclass of Ig-containing proteins, are expressed in a complementary layer-specific fashion in a subset of synaptic partners. We propose that pairs of Dpr/DIP paralogs contribute to layer-specific patterns of synaptic connectivity.


Subject(s)
Drosophila Proteins/metabolism , Immunoglobulins/metabolism , Neurons/metabolism , Receptors, Immunologic/metabolism , Synapses , Animals , Drosophila , Flow Cytometry , Sequence Analysis, RNA , Vision, Ocular
8.
PLoS Genet ; 10(5): e1004380, 2014 May.
Article in English | MEDLINE | ID: mdl-24830287

ABSTRACT

Mature Drosophila sperm are highly polarized cells--on one side is a nearly 2 mm long flagellar tail that comprises most of the cell, while on the other is the sperm head, which carries the gamete's genetic information. The polarization of the sperm cells commences after meiosis is complete and the 64-cell spermatid cyst begins the process of differentiation. The spermatid nuclei cluster to one side of the cyst, while the flagellar axonemes grows from the other. The elongating spermatid bundles are also polarized with respect to the main axis of the testis; the sperm heads are always oriented basally, while the growing tails extend apically. This orientation within the testes is important for transferring the mature sperm into the seminal vesicles. We show here that orienting cyst polarization with respect to the main axis of the testis depends upon atypical Protein Kinase C (aPKC), a factor implicated in polarity decisions in many different biological contexts. When apkc activity is compromised in the male germline, the direction of cyst polarization within this organ is randomized. Significantly, the mechanisms used to spatially restrict apkc activity to the apical side of the spermatid cyst are different from the canonical cross-regulatory interactions between this kinase and other cell polarity proteins that normally orchestrate polarization. We show that the asymmetric accumulation of aPKC protein in the cyst depends on an mRNA localization pathway that is regulated by the Drosophila CPEB protein Orb2. orb2 is required to properly localize and activate the translation of apkc mRNAs in polarizing spermatid cysts. We also show that orb2 functions not only in orienting cyst polarization with respect to the apical-basal axis of the testis, but also in the process of polarization itself. One of the orb2 targets in this process is its own mRNA. Moreover, the proper execution of this orb2 autoregulatory pathway depends upon apkc.


Subject(s)
Drosophila Proteins/genetics , Protein Biosynthesis , Protein Kinase C/genetics , Spermatids/metabolism , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Base Sequence , Drosophila , Male , Meiosis , RNA, Messenger/genetics , Spermatids/cytology
9.
PLoS Genet ; 8(11): e1003079, 2012.
Article in English | MEDLINE | ID: mdl-23209437

ABSTRACT

Cytoplasmic Polyadenylation Element Binding (CPEB) proteins are translational regulators that can either activate or repress translation depending on the target mRNA and the specific biological context. There are two CPEB subfamilies and most animals have one or more genes from each. Drosophila has a single CPEB gene, orb and orb2, from each subfamily. orb expression is only detected at high levels in the germline and has critical functions in oogenesis but not spermatogenesis. By contrast, orb2 is broadly expressed in the soma; and previous studies have revealed important functions in asymmetric cell division, viability, motor function, learning, and memory. Here we show that orb2 is also expressed in the adult male germline and that it has essential functions in programming the progression of spermatogenesis from meiosis through differentiation. Like the translational regulators boule (bol) and off-schedule (ofs), orb2 is required for meiosis and orb2 mutant spermatocytes undergo a prolonged arrest during the meiotic G2-M transition. However, orb2 differs from boule and off-schedule in that this arrest occurs at a later step in meiotic progression after the synthesis of the meiotic regulator twine. orb2 is also required for the orderly differentiation of the spermatids after meiosis is complete. The differentiation defects in orb2 mutants include abnormal elongation of the spermatid flagellar axonemes, a failure in individualization and improper post-meiotic gene expression. Amongst the orb2 differentiation targets are orb and two other mRNAs, which are transcribed post-meiotically and localized to the tip of the flagellar axonemes. Additionally, analysis of a partial loss of function orb2 mutant suggests that the orb2 differentiation phenotypes are independent of the earlier arrest in meiosis.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Meiosis/genetics , Spermatogenesis/genetics , Transcription Factors , mRNA Cleavage and Polyadenylation Factors , Animals , Cell Differentiation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Male , Mutation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
10.
Genetics ; 189(3): 907-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21900268

ABSTRACT

Cytoplasmic polyadenylation element binding (CPEB) proteins bind mRNAs to regulate their localization and translation. While the first CPEBs discovered were germline specific, subsequent studies indicate that CPEBs also function in many somatic tissues including the nervous system. Drosophila has two CPEB family members. One of these, orb, plays a key role in the establishment of polarity axes in the developing egg and early embryo, but has no known somatic functions or expression outside of the germline. Here we characterize the other Drosophila CPEB, orb2. Unlike orb, orb2 mRNA and protein are found throughout development in many different somatic tissues. While orb2 mRNA and protein of maternal origin are distributed uniformly in early embryos, this pattern changes as development proceeds and by midembryogenesis the highest levels are found in the CNS and PNS. In the embryonic CNS, Orb2 appears to be concentrated in cell bodies and mostly absent from the longitudinal and commissural axon tracts. In contrast, in the adult brain, the protein is seen in axonal and dendritic terminals. Lethal effects are observed for both RNAi knockdowns and orb2 mutant alleles while surviving adults display locomotion and behavioral defects. We also show that orb2 funtions in asymmetric division of stem cells and precursor cells during the development of the embryonic nervous system and mesoderm.


Subject(s)
Cell Division , Central Nervous System/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Behavior, Animal/physiology , Cell Line , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , DNA Transposable Elements/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Embryo, Nonmammalian , Embryonic Development/genetics , Embryonic Development/physiology , Female , Genetic Loci/genetics , Larva/genetics , Larva/physiology , Male , Mutation , Ovary/cytology , Ovary/embryology , Ovary/metabolism , Ovum/cytology , Ovum/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Spermatozoa/cytology , Spermatozoa/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/deficiency , mRNA Cleavage and Polyadenylation Factors/genetics
11.
Nat Med ; 15(8): 960-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597504

ABSTRACT

Although the transforming growth factor-beta (TGF-beta) pathway has been implicated in breast cancer metastasis, its in vivo dynamics and temporal-spatial involvement in organ-specific metastasis have not been investigated. Here we engineered a xenograft model system with a conditional control of the TGF-beta-SMAD signaling pathway and a dual-luciferase reporter system for tracing both metastatic burden and TGF-beta signaling activity in vivo. Strong TGF-beta signaling in osteolytic bone lesions is suppressed directly by genetic and pharmacological disruption of the TGF-beta-SMAD pathway and indirectly by inhibition of osteoclast function with bisphosphonates. Notably, disruption of TGF-beta signaling early in metastasis can substantially reduce metastasis burden but becomes less effective when bone lesions are well established. Our in vivo system for real-time manipulation and detection of TGF-beta signaling provides a proof of principle for using similar strategies to analyze the in vivo dynamics of other metastasis-associated signaling pathways and will expedite the development and characterization of therapeutic agents.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Diagnostic Imaging/methods , Transforming Growth Factor beta/pharmacokinetics , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/therapy , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Female , Genetic Vectors , Humans , Mice , Mice, Nude , Models, Biological , Signal Transduction/drug effects , Smad4 Protein/genetics , Smad4 Protein/metabolism , Treatment Outcome , Tumor Cells, Cultured , Validation Studies as Topic , Xenograft Model Antitumor Assays
12.
FEMS Microbiol Lett ; 253(1): 103-9, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16239083

ABSTRACT

A pyocyanin overproducer with insertional inactivation of ptsP gene was isolated from a mini-Mu insertion library in Pseudomonas aeruginosa PA68. The mutation was complemented by a functional ptsP gene in trans. The pyocyanin-overproducing phenotype was also found in a ptsP mutant constructed by gene replacement in the P. aeruginosa PAO1 strain. Reporter plasmids with P(qscR)-lacZ, P(lasI)-lacZ and P(rhlI)-lacZ were constructed and the beta-galactosidase activity in the ptsP mutant/wild-type background was measured. The results showed that lack of Enzyme I(Ntr) (EI(Ntr), encoded by ptsP) decreased transcription from the P(qscR) promoter and increased the activity of the P(lasI) and P(rhlI) promoters. Normally, QscR represses the quorum-sensing LasR-LasI and RhlR-RhlI systems involved in pyocyanin regulation. Our results showed that the ptsP gene has an important role in the regulation of pyocyanin production and that two quorum-sensing systems and their repressor QscR are involved in this regulation.


Subject(s)
Genes, Bacterial , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pyocyanine/biosynthesis , Base Sequence , DNA, Bacterial/genetics , Gene Expression , Genes, Reporter , Genetic Complementation Test , Kinetics , Lac Operon , Mutagenesis, Insertional , Phenotype , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...