Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Cell ; 156(6): 1259-1273, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24630726

ABSTRACT

Members of the SWI/SNF chromatin-remodeling complex are among the most frequently mutated genes in human cancer, but how they suppress tumorigenesis is currently unclear. Here, we use Drosophila neuroblasts to demonstrate that the SWI/SNF component Osa (ARID1) prevents tumorigenesis by ensuring correct lineage progression in stem cell lineages. We show that Osa induces a transcriptional program in the transit-amplifying population that initiates temporal patterning, limits self-renewal, and prevents dedifferentiation. We identify the Prdm protein Hamlet as a key component of this program. Hamlet is directly induced by Osa and regulates the progression of progenitors through distinct transcriptional states to limit the number of transit-amplifying divisions. Our data provide a mechanistic explanation for the widespread tumor suppressor activity of SWI/SNF. Because the Hamlet homologs Evi1 and Prdm16 are frequently mutated in cancer, this mechanism could well be conserved in human stem cell lineages. PAPERCLIP:


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Brain/cytology , Brain/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic
2.
Proc Natl Acad Sci U S A ; 116(49): 24830-24839, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31748267

ABSTRACT

Severe locomotor impairment is a common phenotype of neurodegenerative disorders such as Parkinson's disease (PD). Drosophila models of PD, studied for more than a decade, have helped in understanding the interaction between various genetic factors, such as parkin and PINK1, in this disease. To characterize locomotor behavioral phenotypes for these genes, fly climbing assays have been widely used. While these simple current assays for locomotor defects in Drosophila mutants measure some locomotor phenotypes well, it is possible that detection of subtle changes in behavior is important to understand the manifestation of locomotor disorders. We introduce a climbing behavior assay which provides such fine-scale behavioral data and tests this proposition for the Drosophila model. We use this inexpensive, fully automated assay to quantitatively characterize the climbing behavior at high parametric resolution in 3 contexts. First, we characterize wild-type flies and uncover a hitherto unknown sexual dimorphism in climbing behavior. Second, we study climbing behavior of heterozygous mutants of genes implicated in the fly PD model and reveal previously unreported prominent locomotor defects in some of these heterozygous fly lines. Finally, we study locomotor defects in a homozygous proprioceptory mutation (Trp-γ1 ) known to affect fine motor control in Drosophila Moreover, we identify aberrant geotactic behavior in Trp-γ1 mutants, thereby opening up a finer assay for geotaxis and its genetic basis. Our assay is therefore a cost-effective, general tool for measuring locomotor behaviors of wild-type and mutant flies in fine detail and can reveal subtle motor defects.


Subject(s)
Behavior Observation Techniques/methods , Behavior, Animal/physiology , Locomotion/genetics , Parkinson Disease/genetics , Proprioception/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Heterozygote , Homozygote , Humans , Male , Parkinson Disease/physiopathology , Protein Serine-Threonine Kinases/genetics , Sensitivity and Specificity , Sex Characteristics , Transient Receptor Potential Channels/genetics , Ubiquitin-Protein Ligases/genetics
3.
Proc Natl Acad Sci U S A ; 115(9): E2115-E2124, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440493

ABSTRACT

Walking is a complex rhythmic locomotor behavior generated by sequential and periodical contraction of muscles essential for coordinated control of movements of legs and leg joints. Studies of walking in vertebrates and invertebrates have revealed that premotor neural circuitry generates a basic rhythmic pattern that is sculpted by sensory feedback and ultimately controls the amplitude and phase of the motor output to leg muscles. However, the identity and functional roles of the premotor interneurons that directly control leg motoneuron activity are poorly understood. Here we take advantage of the powerful genetic methodology available in Drosophila to investigate the role of premotor inhibition in walking by genetically suppressing inhibitory input to leg motoneurons. For this, we have developed an algorithm for automated analysis of leg motion to characterize the walking parameters of wild-type flies from high-speed video recordings. Further, we use genetic reagents for targeted RNAi knockdown of inhibitory neurotransmitter receptors in leg motoneurons together with quantitative analysis of resulting changes in leg movement parameters in freely walking Drosophila Our findings indicate that targeted down-regulation of the GABAA receptor Rdl (Resistance to Dieldrin) in leg motoneurons results in a dramatic reduction of walking speed and step length without the loss of general leg coordination during locomotion. Genetically restricting the knockdown to the adult stage and subsets of motoneurons yields qualitatively identical results. Taken together, these findings identify GABAergic premotor inhibition of motoneurons as an important determinant of correctly coordinated leg movements and speed of walking in freely behaving Drosophila.


Subject(s)
Drosophila/physiology , Locomotion/physiology , Motor Neurons/physiology , Walking/physiology , Algorithms , Animals , Animals, Genetically Modified , Electromyography , Electronic Data Processing , Extremities/physiology , Feedback, Sensory , Immunohistochemistry , Interneurons/physiology , Introns , Male , Microscopy, Confocal , Neurotransmitter Agents/physiology , Periodicity , Phenotype , RNA Interference , Signal Processing, Computer-Assisted , Video Recording
4.
Development ; 144(21): 3932-3945, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28935704

ABSTRACT

Stem cells need to balance self-renewal and differentiation for correct tissue development and homeostasis. Defects in this balance can lead to developmental defects or tumor formation. In recent years, mRNA splicing has emerged as an important mechanism regulating cell fate decisions. Here we address the role of the evolutionarily conserved splicing co-factor Barricade (Barc)/Tat-SF1/CUS2 in Drosophila neural stem cell (neuroblast) lineage formation. We show that Barc is required for the generation of neurons during Drosophila brain development by ensuring correct neural progenitor proliferation and differentiation. Barc associates with components of the U2 small nuclear ribonucleoprotein (snRNP) complex, and its depletion causes alternative splicing in the form of intron retention in a subset of genes. Using bioinformatics analysis and a cell culture-based splicing assay, we found that Barc-dependent introns share three major traits: they are short, GC rich and have weak 3' splice sites. Our results show that Barc, together with the U2 snRNP complex, plays an important role in regulating neural stem cell lineage progression during brain development and facilitates correct splicing of a subset of introns.


Subject(s)
Cell Cycle , Cell Lineage , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transcription Factors/metabolism , Alternative Splicing/genetics , Animals , Base Composition/genetics , Base Sequence , Body Patterning/genetics , Brain/anatomy & histology , Cell Count , Cell Proliferation , Clone Cells , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Knockdown Techniques , Introns/genetics , Mice , Models, Biological , Mutation/genetics , Neurons/cytology , Neurons/metabolism , Phenotype , Protein Binding , RNA Interference , RNA Splice Sites/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Time Factors
5.
Bioessays ; 40(10): e1800107, 2018 10.
Article in English | MEDLINE | ID: mdl-30151860

ABSTRACT

The acoel worm Symsagittifera roscoffensis, an early offshoot of the Bilateria and the only well-studied marine acoel that lives in a photosymbiotic relationship, exhibits a centralized nervous system, brain regeneration, and a wide repertoire of complex behaviors such as circatidal rhythmicity, photo/geotaxis, and social interactions. While this animal can be collected by the thousands and is studied historically, significant progress is made over the last decade to develop it as an emerging marine model. The authors here present the feasibility of culturing it in the laboratory and describe the progress made on different areas, including genomic and tissue architectures, highlighting the associated challenges. In light of these developments, and on the ability to access abundant synchronized embryos, the authors put forward S. roscoffensis as a marine system to revisit questions in the areas of photosymbiosis, regeneration, chronobiology, and the study of complex behaviors from a molecular and evolutionary perspective.


Subject(s)
Brain/physiology , Platyhelminths/physiology , Regeneration/physiology , Animals , Aquatic Organisms , Behavior, Animal , Brain/cytology , Chronobiology Phenomena , Circadian Rhythm/genetics , Microalgae/physiology , Microbiota/physiology , Sulfonium Compounds/metabolism , Symbiosis , Totipotent Stem Cells/physiology
6.
Semin Cell Dev Biol ; 72: 56-66, 2017 12.
Article in English | MEDLINE | ID: mdl-29146144

ABSTRACT

Myogenesis is a highly orchestrated, complex developmental process by which cell lineages that are mesodermal in origin generate differentiated multinucleate muscle cells as a final product. Considerable insight into the process of myogenesis has been obtained for the embryonic development of the larval muscles of Drosophila. More recently, the postembryonic development of the muscles of the adult fly has become a focus of experimental investigation of myogenesis since specific flight muscles of the fly manifest remarkable similarities to vertebrate muscles in their development and organization. In this review, we catalog some of the milestones in the study of myogenesis in the large adult-specific flight muscles of Drosophila. The identification of mesoderm-derived muscle stem cell lineages, the characterization of the symmetric and asymmetric divisions through which they produce adult-specific myoblasts, the multifaceted processes of myoblast fusion, and the unexpected discovery of quiescent satellite cells that can be activated by injury are discussed. Moreover, the finding that all of these processes incorporate a plethora of signaling interactions with other myogenic cells and with niche-like neighboring tissue is considered. Finally, we briefly point out possible future developments in the area of Drosophila myogenesis that may lead to of new avenues of genetic research into the roles of muscle stem cells in development, disease and aging.


Subject(s)
Drosophila/genetics , Gene Expression Regulation, Developmental , Muscle Development/genetics , Muscles/metabolism , Animals , Drosophila/growth & development , Models, Genetic , Morphogenesis/genetics , Muscle Fibers, Skeletal/metabolism , Muscles/physiology , Myoblasts/metabolism , Regeneration/genetics
7.
Article in English | MEDLINE | ID: mdl-28831545

ABSTRACT

In the 21st century, neurobiological studies focused on the insect brain are revealing unprecedented insight into the molecular, cellular, developmental, and circuit aspects of brain organization and function, notably in the genetic model system of Drosophila melanogaster. Underlying this accelerating progress in understanding the insect brain is a century-long history of ground breaking experimental investigation, methodological advance, and conceptual insight catalyzed by the integration of two emerging research fields, neuroscience and genetics. This review traces some of the key early steps in this remarkable historical scientific adventure of exploring the brain of "these apparently humble representatives of life".


Subject(s)
Insecta/genetics , Insecta/physiology , Models, Animal , Animals , Brain/physiology
8.
Dev Biol ; 390(2): 102-15, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24713419

ABSTRACT

The central brain of Drosophila consists of the supraesophageal ganglion (SPG) and the subesophageal ganglion (SEG), both of which are generated by neural stem cell-like neuroblasts during embryonic and postembryonic development. Considerable information has been obtained on postembryonic development of the neuroblasts and their lineages in the SPG. In contrast, very little is known about neuroblasts, neural lineages, or any other aspect of the postembryonic development in the SEG. Here we characterize the neuroanatomy of the larval SEG in terms of tracts, commissures, and other landmark features as compared to a thoracic ganglion. We then use clonal MARCM labeling to identify all adult-specific neuroblast lineages in the late larval SEG and find a surprisingly small number of neuroblast lineages, 13 paired and one unpaired. The Hox genes Dfd, Scr, and Antp are expressed in a lineage-specific manner in these lineages during postembryonic development. Hox gene loss-of-function causes lineage-specific defects in axonal targeting and reduction in neural cell numbers. Moreover, it results in the formation of novel ectopic neuroblast lineages. Apoptosis block also results in ectopic lineages suggesting that Hox genes are required for lineage-specific termination of proliferation through programmed cell death. Taken together, our findings show that postembryonic development in the SEG is mediated by a surprisingly small set of identified lineages and requires lineage-specific Hox gene action to ensure the correct formation of adult-specific neurons in the Drosophila brain.


Subject(s)
Brain/growth & development , Cell Lineage/physiology , Drosophila/growth & development , Ganglia, Invertebrate/growth & development , Gene Expression Regulation, Developmental/physiology , Genes, Homeobox/physiology , Neural Stem Cells/physiology , Animals , Brain/metabolism , Drosophila/genetics , Ganglia, Invertebrate/metabolism , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox/genetics , Immunohistochemistry , Microscopy, Confocal
9.
Cell Tissue Res ; 359(1): 33-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24902665

ABSTRACT

The neural stem cells of Drosophila, called neuroblasts, have the ability to self-renew and at the same time produce many different types of neurons and glial cells. In the central brain and ventral ganglia, neuroblasts are specified and delaminate from the neuroectoderm during embryonic development under the control of proneural and neurogenic genes. In contrast, in the optic lobes, neuroepithelial cells are transformed into neuroblasts postembryonically by a spatial wave of proneural gene expression. Central brain and ventral nerve cord neuroblasts manifest a short embryonic proliferation period followed by a stage of quiescence and then undergo a prolonged postembryonic proliferation period during which most of the differentiated neurons of the adult CNS are generated. While most neuroblasts belong to a type I class that produces neuronal lineages through non-self-renewing ganglion mother cells, a small subset of type II neuroblasts generates exceptionally large neuronal lineages through self-renewing intermediate progenitor cells that have a transit amplifying function. All neuroblasts in the CNS generate their neural progeny through an asymmetric cell division mode in which the interplay of apical complex and basal complex molecules in the mitotically active progenitor results in the segregation of cell fate determinants into the smaller more differentiated daughter cell. Defects in this molecular control of asymmetric cell division in neuroblasts can result in brain tumor formation. Proliferating neuroblast lineages in the developing CNS utilize transcription factor cascades as a generic mechanism for temporal patterning and birth order-dependent determination of differential neural cell fate. This contributes to the generation of a remarkable diversity of cell types in the developing CNS from a surprisingly small set of neural stem cell-like precursors.


Subject(s)
Cell Differentiation , Drosophila melanogaster/cytology , Neural Stem Cells/cytology , Animals , Cell Proliferation , Models, Biological , Neurogenesis
10.
Dev Biol ; 379(2): 182-94, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23628691

ABSTRACT

The neurons and glial cells of the Drosophila brain are generated by neural stem cell-like progenitors during two developmental phases, one short embryonic phase and one more prolonged postembryonic phase. Like the bulk of the adult-specific neurons, most of glial cells found in the adult central brain are generated postembryonically. Five of the neural stem cell-like progenitors that give rise to glial cells during postembryonic brain development have been identified as type II neuroglioblasts that generate neural and glial progeny through transient amplifying INPs. Here we identify DL1 as a novel multipotent neuroglial progenitor in the central brain and show that this type II neuroblast not only gives rise to neurons that innervate the central complex but also to glial cells that contribute exclusively to the optic lobe. Immediately following their generation in the central brain during the second half of larval development, these DL1 lineage-derived glia migrate into the developing optic lobe, where they differentiate into three identified types of optic lobe glial cells, inner chiasm glia, outer chiasm glia and cortex glia. Taken together, these findings reveal an unexpected central brain origin of optic lobe glial cells and central complex interneurons from one and the same type II neuroglioblast.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Drosophila melanogaster/growth & development , Neural Stem Cells/physiology , Neuroglia/physiology , Optic Lobe, Nonmammalian/growth & development , Animals , Cell Movement/physiology , Immunohistochemistry , Larva/growth & development , Microscopy, Fluorescence , Multipotent Stem Cells/physiology , Optic Lobe, Nonmammalian/cytology
11.
Dev Biol ; 381(1): 17-27, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23810656

ABSTRACT

An important role in olfactory system development is played by transcription factors which act in sensory neurons or in their interneuron targets as cell autonomous regulators of downstream effectors such as cell surface molecules and signalling systems that control neuronal identity and process guidance. Some of these transcriptional regulators have been characterized in detail in the development of the neural elements that innervate the antennal lobe in the olfactory system of Drosophila. Here we identify the zinc finger transcription factor Jing as a cell autonomously acting transcriptional regulator that is required both for dendrite targeting of projection neurons and local interneurons as well as for axonal targeting of olfactory sensory neurons in Drosophila olfactory system development. Immunocytochemical analysis shows that Jing is widely expressed in the neural cells during postembryonic development. MARCM-based clonal analysis of projection neuron and local interneuron lineages reveals a requirement for Jing in dendrite targeting; Jing loss-of-function results in loss of innervation in specific glomeruli, ectopic innervation of inappropriate glomeruli, aberrant profuse dendrite arborisation throughout the antennal lobe, as well as mistargeting to other parts of the CNS. ey-FLP-based MARCM analysis of olfactory sensory neurons reveals an additional requirement for Jing in axonal targeting; mutational inactivation of Jing causes specific mistargeting of some olfactory sensory neuron axons to the DA1 glomerulus, reduction of targeting to other glomeruli, as well as aberrant stalling of axons in the antennal lobe. Taken together, these findings indicate that Jing acts as a key transcriptional control element in wiring of the circuitry in the developing olfactory sensory system in Drosophila.


Subject(s)
Arthropod Antennae/metabolism , Axons/metabolism , Dendrites/metabolism , Drosophila Proteins/genetics , Nuclear Proteins/genetics , Olfactory Pathways/embryology , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Cell Lineage , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Interneurons/metabolism , Mutation , Nuclear Proteins/metabolism , Olfactory Pathways/metabolism , Olfactory Receptor Neurons/embryology , Transcription Factors/metabolism , Zinc Fingers
12.
Dev Biol ; 373(2): 322-37, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23149077

ABSTRACT

The complete neuronal repertoire of the central brain of Drosophila originates from only approximately 100 pairs of neural stem cells, or neuroblasts. Each neuroblast produces a highly stereotyped lineage of neurons which innervate specific compartments of the brain. Neuroblasts undergo two rounds of mitotic activity: embryonic divisions produce lineages of primary neurons that build the larval nervous system; after a brief quiescence, the neuroblasts go through a second round of divisions in larval stage to produce secondary neurons which are integrated into the adult nervous system. Here we investigate the lineages that are associated with the larval antennal lobe, one of the most widely studied neuronal systems in fly. We find that the same five neuroblasts responsible for the adult antennal lobe also produce the antennal lobe of the larval brain. However, there are notable differences in the composition of larval (primary) lineages and their adult (secondary) counterparts. Significantly, in the adult, two lineages (lNB/BAlc and adNB/BAmv3) produce uniglomerular projection neurons connecting the antennal lobe with the mushroom body and lateral horn; another lineage, vNB/BAla1, generates multiglomerular neurons reaching the lateral horn directly. lNB/BAlc, as well as a fourth lineage, vlNB/BAla2, generate a diversity of local interneurons. We describe a fifth, previously unknown lineage, BAlp4, which connects the posterior part of the antennal lobe and the neighboring tritocerebrum (gustatory center) with a higher brain center located adjacent to the mushroom body. In the larva, only one of these lineages, adNB/BAmv3, generates all uniglomerular projection neurons. Also as in the adult, lNB/BAlc and vlNB/BAla2 produce local interneurons which, in terms of diversity in architecture and transmitter expression, resemble their adult counterparts. In addition, lineages lNB/BAlc and vNB/BAla1, as well as the newly described BAlp4, form numerous types of projection neurons which along the same major axon pathways (antennal tracts) used by the antennal projection neurons, but which form connections that include regions outside the "classical" olfactory circuit triad antennal lobe-mushroom body-lateral horn. Our work will benefit functional studies of the larval olfactory circuit, and shed light on the relationship between larval and adult neurons.


Subject(s)
Cell Lineage , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Neurons/cytology , Olfactory Pathways/cytology , Animals , Arthropod Antennae/cytology , Brain/cytology , Interneurons/cytology , Interneurons/metabolism , Larva/cytology , Larva/growth & development , Olfactory Pathways/growth & development , Pupa/cytology
13.
J Neurogenet ; 28(3-4): 181-9, 2014.
Article in English | MEDLINE | ID: mdl-24766377

ABSTRACT

Neuroblasts, the neural stem cells in Drosophila, generate the complex neural structure of the central nervous system. Significant progress has been made in understanding the mechanisms regulating the self-renewal, proliferation, and differentiation in Drosophila neuroblast lineages. Deregulation of these mechanisms can lead to severe developmental defects and the formation of malignant brain tumors. Here, the authors review the molecular genetics of Drosophila neuroblasts and discuss some recent advances in stem cell and cancer biology using this model system.


Subject(s)
Brain/growth & development , Neoplasms/pathology , Neural Stem Cells/cytology , Animals , Brain/pathology , Cell Division , Cell Lineage , Drosophila
14.
Dev Genes Evol ; 223(3): 149-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23143292

ABSTRACT

Flatworms are classically considered to represent the simplest organizational form of all living bilaterians with a true central nervous system. Based on their simple body plans, all flatworms have been traditionally grouped together in a single phylum at the base of the bilaterians. Current molecular phylogenomic studies now split the flatworms into two widely separated clades, the acoelomorph flatworms and the platyhelminth flatworms, such that the last common ancestor of both clades corresponds to the urbilaterian ancestor of all bilaterian animals. Remarkably, recent comparative neuroanatomical analyses of acoelomorphs and platyhelminths show that both of these flatworm groups have complex anterior brains with surprisingly similar basic neuroarchitectures. Taken together, these findings imply that fundamental neuroanatomical features of the brain in the two separate flatworm groups are likely to be primitive and derived from the urbilaterian brain.


Subject(s)
Brain/anatomy & histology , Platyhelminths/classification , Animals , Phylogeny
15.
Development ; 137(21): 3687-95, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20940227

ABSTRACT

In Drosophila, the cephalic gap gene empty spiracles plays key roles in embryonic patterning of the peripheral and central nervous system. During postembryonic development, it is involved in the development of central olfactory circuitry in the antennal lobe of the adult. However, its possible role in the postembryonic development of peripheral olfactory sense organs has not been investigated. Here, we show that empty spiracles acts in a subset of precursors that generate the olfactory sense organs of the adult antenna. All empty spiracles-expressing precursor cells co-express the proneural gene amos and the early patterning gene lozenge. Moreover, the expression of empty spiracles in these precursor cells is dependent on both amos and lozenge. Functional analysis reveals two distinct roles of empty spiracles in the development of olfactory sense organs. Genetic interaction studies in a lozenge-sensitized background uncover a requirement of empty spiracles in the formation of trichoid and basiconic olfactory sensilla. MARCM-based clonal mutant analysis reveals an additional role during axonal targeting of olfactory sensory neurons to glomeruli within the antennal lobe. Our findings on empty spiracles action in olfactory sense organ development complement previous studies that demonstrate its requirement in olfactory interneurons and, taken together with studies on the murine homologs of empty spiracles, suggest that conserved molecular genetic programs might be responsible for the formation of both peripheral and central olfactory circuitry in insects and mammals.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Olfactory Pathways/embryology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Male , Models, Biological , Olfactory Pathways/growth & development , Olfactory Pathways/metabolism , Olfactory Receptor Neurons/embryology , Olfactory Receptor Neurons/metabolism , Sense Organs/embryology , Sense Organs/metabolism , Smell/genetics , Stem Cells/metabolism , Stem Cells/physiology
16.
Arch Biochem Biophys ; 534(1-2): 38-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22906721

ABSTRACT

The fruit fly, Drosophila melanogaster, has proved to be a useful model organism for studying the biology of neural stem cells. Notably, significant progress has been made in identifying the molecular mechanisms that regulate the asymmetric cell divisions of the neural stem cell-like neuroblasts during brain development. Recently, the emerging technology of genome-wide transgenic RNA interference (RNAi), which makes it possible to analyze complicated developmental processes in a targeted, tissue-specific way, has been used for the analysis of gene function in Drosophila neuroblasts. Here, we review the key molecular mechanisms that regulate the asymmetric cell divisions of neuroblasts during brain development in Drosophila. We then summarize recent genome-wide transgenic RNAi screens in Drosophila and report on the identification of new regulators and gene networks that are required in balancing neuroblast self-renewal and differentiation.


Subject(s)
Cell Differentiation , Drosophila melanogaster/cytology , Gene Expression Regulation, Developmental , Genes, Insect , Neural Stem Cells/cytology , RNA Interference , Animals , Asymmetric Cell Division , Brain/cytology , Brain/embryology , Brain/metabolism , Cell Proliferation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Regulatory Networks , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism
17.
Front Zool ; 10(1): 19, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23587092

ABSTRACT

INTRODUCTION: Loricifera is a group of small, marine animals, with undetermined phylogenetic relationships within Ecdysozoa (molting protostome animals). Despite their well-known external morphology, data on the internal anatomy of loriciferans are still incomplete. Aiming to increase the knowledge of this enigmatic phylum, we reconstruct for the first time the three-dimensional myoanatomy of loriciferans. Adult Nanaloricus sp. and the Higgins larva of Armorloricus elegans were investigated with cytochemical labeling techniques and CLSM. We discuss our findings with reference to other loriciferan species and recently established phylogenies. RESULTS: The somatic musculature of both adult and larval stages is very complex and includes several muscles arranged in three orientations: circular, transverse and longitudinal. In adult Nanaloricus sp., the introvert is characterized by a net-like muscular arrangement, which is composed of five thin circular fibers crossed by several (up to 30) thin longitudinal fibers with bifurcated anterior ends. Two sets of muscles surround the pre-pharyngeal armature: 6 buccal tube retractors arranged 3 × 2 in a conical shaped structure, and 8 mouth cone retractors. Additionally, a thick, circular muscle marks the neck region and a putative anal sphincter is the posteriormost myoanatomical feature. In the Higgins larva of A. elegans, two circular muscles are distinguished anteriorly in the introvert: a dorsal semicircular fiber and a thin ring muscle. The posteriormost region of the body is characterized by an anal sphincter and a triangular muscle. CONCLUSIONS: Based on the currently available knowledge, the myoanatomical bodyplan of adult loriciferans includes: (i) 8 mouth cone retractors, (ii) a pharynx bulb composed of transversal fibers arranged radially, (iii) circular muscles of the head and neck, (iv) internal muscles of the spinoscalids, (v) longitudinal muscles spanning all body regions, and (vi) transverse (circular) muscles in the abdomen. Concerning the Higgins larva, the muscle subsets assigned to its myoanatomical ground pattern are the (i) longitudinal retractors of the mouth cone, introvert, and abdomen, (ii) abdominal transverse muscles, and (iii) a pharynx bulb composed of transverse, radial fibers. In a comparison with phyla traditionally regarded as phylogenetically close, our data show that the overall myoanatomy of Loricifera is more similar to Kinorhyncha and Nematomorpha than to Priapulida. However, the head musculature of all these groups is very similar, which supports homology of their introverts and head morphology.

18.
Dev Biol ; 356(2): 553-65, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21708145

ABSTRACT

The neural stem cells that give rise to the neural lineages of the brain can generate their progeny directly or through transit amplifying intermediate neural progenitor cells (INPs). The INP-producing neural stem cells in Drosophila are called type II neuroblasts, and their neural progeny innervate the central complex, a prominent integrative brain center. Here we use genetic lineage tracing and clonal analysis to show that the INPs of these type II neuroblast lineages give rise to glial cells as well as neurons during postembryonic brain development. Our data indicate that two main types of INP lineages are generated, namely mixed neuronal/glial lineages and neuronal lineages. Genetic loss-of-function and gain-of-function experiments show that the gcm gene is necessary and sufficient for gliogenesis in these lineages. The INP-derived glial cells, like the INP-derived neuronal cells, make major contributions to the central complex. In postembryonic development, these INP-derived glial cells surround the entire developing central complex neuropile, and once the major compartments of the central complex are formed, they also delimit each of these compartments. During this process, the number of these glial cells in the central complex is increased markedly through local proliferation based on glial cell mitosis. Taken together, these findings uncover a novel and complex form of neurogliogenesis in Drosophila involving transit amplifying intermediate progenitors. Moreover, they indicate that type II neuroblasts are remarkably multipotent neural stem cells that can generate both the neuronal and the glial progeny that make major contributions to one and the same complex brain structure.


Subject(s)
Brain/embryology , Drosophila melanogaster/embryology , Multipotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Neuroglia/cytology , Animals , Cell Lineage , Cell Proliferation
19.
Development ; 136(20): 3433-42, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19762424

ABSTRACT

The Drosophila central brain is composed of thousands of neurons that derive from approximately 100 neuroblasts per hemisphere. Functional circuits in the brain require precise neuronal wiring and tight control of neuronal numbers. How this accurate control of neuronal numbers is achieved during neural development is largely unclear. Specifically, the role of programmed cell death in control of cell numbers has not been studied in the central brain neuroblast lineages. Here, we focus on four postembryonic neuroblast lineages in the central brain identified on the basis that they express the homeobox gene engrailed (en). For each lineage, we determine the total number of adult-specific neurons generated as well as number and pattern of en-expressing cells. We then demonstrate that programmed cell death has a pronounced effect on the number of cells in the four lineages; approximately half of the immature adult-specific neurons in three of the four lineages are eliminated by cell death during postembryonic development. Moreover, we show that programmed cell death selectively affects en-positive versus en-negative cells in a lineage-specific manner and, thus, controls the relative number of en-expressing neurons in each lineage. Furthermore, we provide evidence that Notch signaling is involved in the regulation of en expression. Based on our findings, we conclude that lineage-specific programmed cell death plays a prominent role in the generation of neuronal number and lineage diversity in the Drosophila brain.


Subject(s)
Cell Lineage , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Signal Transduction , Animals , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Death , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Dev Biol ; 340(1): 125-33, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20045685

ABSTRACT

The ems/Emx genes encode homeodomain transcription factors that have conserved actions in anterior embryonic patterning in bilaterian animals ranging from insects to mammals. Recently, genes of the ems/Emx family have been identified in cnidarians raising the possibility that some of their developmental functions might be conserved throughout the Eumetazoa. To determine to what extent functions of a cnidarian ems/Emx protein have been retained across phyla, we carried out cross-phylum rescue expression experiments in which the coral Acropora emx-Am gene was misexpressed in Drosophila ems mutants. Our findings demonstrate that coral emx-Am can substitute for fly ems in embryonic head development and rescue the open head defect and the loss of segmental engrailed expression domains in Drosophila ems mutants. In contrast, the coral emx-Am gene can not substitute for fly ems in embryonic brain development. Even when a hexapeptide motif of the type present in the Drosophila ems gene is inserted into the coral emx-Am gene, rescue of the developmental brain defects in fly ems mutants fails. These findings have implications for understanding the evolutionary origins of head versus brain patterning mechanisms.


Subject(s)
Drosophila/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Anthozoa/genetics , Anthozoa/metabolism , Body Patterning/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Homeodomain Proteins/metabolism , Microscopy, Confocal , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL