Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Dev Biol ; 475: 165-180, 2021 07.
Article in English | MEDLINE | ID: mdl-32017903

ABSTRACT

Complex nervous systems have a modular architecture, whereby reiterative groups of neurons ("modules") that share certain structural and functional properties are integrated into large neural circuits. Neurons develop from proliferating progenitor cells that, based on their location and time of appearance, are defined by certain genetic programs. Given that genes expressed by a given progenitor play a fundamental role in determining the properties of its lineage (i.e., the neurons descended from that progenitor), one efficient developmental strategy would be to have lineages give rise to the structural modules of the mature nervous system. It is clear that this strategy plays an important role in neural development of many invertebrate animals, notably insects, where the availability of genetic techniques has made it possible to analyze the precise relationship between neuronal origin and differentiation since several decades. Similar techniques, developed more recently in the vertebrate field, reveal that functional modules of the mammalian cerebral cortex are also likely products of developmentally defined lineages. We will review studies that relate cell lineage to circuitry and function from a comparative developmental perspective, aiming at enhancing our understanding of neural progenitors and their lineages, and translating findings acquired in different model systems into a common conceptual framework.


Subject(s)
Cell Lineage/physiology , Nerve Net/cytology , Neurons/cytology , Animals , Brain/cytology , Cell Differentiation , Cell Lineage/genetics , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental , Humans , Nerve Net/metabolism , Nerve Net/physiology , Nervous System/cytology , Neurons/metabolism , Neurons/physiology , Stem Cells/cytology
2.
Front Neural Circuits ; 12: 103, 2018.
Article in English | MEDLINE | ID: mdl-30546298

ABSTRACT

The central complex (CX) is a midline-situated collection of neuropil compartments in the arthropod central brain, implicated in higher-order processes such as goal-directed navigation. Here, we provide a systematic genetic-neuroanatomical analysis of the ellipsoid body (EB), a compartment which represents a major afferent portal of the Drosophila CX. The neuropil volume of the EB, along with its prominent input compartment, called the bulb, is subdivided into precisely tessellated domains, distinguishable based on intensity of the global marker DN-cadherin. EB tangential elements (so-called ring neurons), most of which are derived from the DALv2 neuroblast lineage, predominantly interconnect the bulb and EB domains in a topographically organized fashion. Using the DN-cadherin domains as a framework, we first characterized this connectivity by Gal4 driver lines expressed in different DALv2 ring neuron (R-neuron) subclasses. We identified 11 subclasses, 6 of which correspond to previously described projection patterns, and 5 novel patterns. These subclasses both spatially (based on EB innervation pattern) and numerically (cell counts) summate to the total EB volume and R-neuron cell number, suggesting that our compilation of R-neuron subclasses approaches completion. EB columnar elements, as well as non-DALv2 derived extrinsic ring neurons (ExR-neurons), were also incorporated into this anatomical framework. Finally, we addressed the connectivity between R-neurons and their targets, using the anterograde trans-synaptic labeling method, trans-Tango. This study demonstrates putative interactions of R-neuron subclasses and reveals general principles of information flow within the EB network. Our work will facilitate the generation and testing of hypotheses regarding circuit interactions within the EB and the rest of the CX.


Subject(s)
Nerve Net/physiology , Nerve Net/ultrastructure , Neuronal Plasticity/physiology , Neuropil/physiology , Neuropil/ultrastructure , Animals , Animals, Genetically Modified , Drosophila , Female , Nerve Net/cytology , Neurons/physiology , Neurons/ultrastructure
3.
J Comp Neurol ; 526(1): 6-32, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28730682

ABSTRACT

The subesophageal zone (SEZ) of the Drosophila brain houses the circuitry underlying feeding behavior and is involved in many other aspects of sensory processing and locomotor control. Formed by the merging of four neuromeres, the internal architecture of the SEZ can be best understood by identifying segmentally reiterated landmarks emerging in the embryo and larva, and following the gradual changes by which these landmarks become integrated into the mature SEZ during metamorphosis. In previous works, the system of longitudinal fibers (connectives) and transverse axons (commissures) has been used as a scaffold that provides internal landmarks for the neuromeres of the larval ventral nerve cord. We have extended the analysis of this scaffold to the SEZ and, in addition, reconstructed the tracts formed by lineages and nerves in relationship to the connectives and commissures. As a result, we establish reliable criteria that define boundaries between the four neuromeres (tritocerebrum, mandibular neuromere, maxillary neuromere, labial neuromere) of the SEZ at all stages of development. Fascicles and lineage tracts also demarcate seven columnar neuropil domains (ventromedial, ventro-lateral, centromedial, central, centrolateral, dorsomedial, dorsolateral) identifiable throughout development. These anatomical subdivisions, presented in the form of an atlas including confocal sections and 3D digital models for the larval, pupal and adult stage, allowed us to describe the morphogenetic changes shaping the adult SEZ. Finally, we mapped MARCM-labeled clones of all secondary lineages of the SEZ to the newly established neuropil subdivisions. Our work will facilitate future studies of function and comparative anatomy of the SEZ.


Subject(s)
Brain , Cell Lineage/physiology , Drosophila , Metamorphosis, Biological , Neurons/cytology , Animals , Animals, Genetically Modified , Brain/anatomy & histology , Brain/embryology , Brain/growth & development , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila/anatomy & histology , Drosophila/embryology , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Larva , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Neurons/metabolism , Neuropil/metabolism
4.
J Comp Neurol ; 525(16): 3458-3475, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28675433

ABSTRACT

The anterior visual pathway (AVP) conducts visual information from the medulla of the optic lobe via the anterior optic tubercle (AOTU) and bulb (BU) to the ellipsoid body (EB) of the central complex. The anatomically defined neuron classes connecting the AOTU, BU, and EB represent discrete lineages, genetically and developmentally specified sets of cells derived from common progenitors (Omoto et al., Current Biology, 27, 1098-1110, 2017). In this article, we have analyzed the formation of the AVP from early larval to adult stages. The immature fiber tracts of the AVP, formed by secondary neurons of lineages DALcl1/2 and DALv2, assemble into structurally distinct primordia of the AOTU, BU, and EB within the late larval brain. During the early pupal period (P6-P48) these primordia grow in size and differentiate into the definitive subcompartments of the AOTU, BU, and EB. The primordium of the EB has a complex composition. DALv2 neurons form the anterior EB primordium, which starts out as a bilateral structure, then crosses the midline between P6 and P12, and subsequently bends to adopt the ring shape of the mature EB. Columnar neurons of the central complex, generated by the type II lineages DM1-4, form the posterior EB primordium. Starting out as an integral part of the fan-shaped body primordium, the posterior EB primordium moves forward and merges with the anterior EB primordium. We document the extension of neuropil glia around the nascent EB and BU, and analyze the relationship of primary and secondary neurons of the AVP lineages.


Subject(s)
Brain/physiology , Cell Lineage , Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Visual Pathways/physiology , Animals , Animals, Genetically Modified , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Larva , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Neurons/cytology , Neuropil/metabolism , Neuropil/physiology , Optic Nerve/physiology , Pupa , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Curr Biol ; 27(8): 1098-1110, 2017 Apr 24.
Article in English | MEDLINE | ID: mdl-28366740

ABSTRACT

The Drosophila central brain consists of stereotyped neural lineages, developmental-structural units of macrocircuitry formed by the sibling neurons of single progenitors called neuroblasts. We demonstrate that the lineage principle guides the connectivity and function of neurons, providing input to the central complex, a collection of neuropil compartments important for visually guided behaviors. One of these compartments is the ellipsoid body (EB), a structure formed largely by the axons of ring (R) neurons, all of which are generated by a single lineage, DALv2. Two further lineages, DALcl1 and DALcl2, produce neurons that connect the anterior optic tubercle, a central brain visual center, with R neurons. Finally, DALcl1/2 receive input from visual projection neurons of the optic lobe medulla, completing a three-legged circuit that we call the anterior visual pathway (AVP). The AVP bears a fundamental resemblance to the sky-compass pathway, a visual navigation circuit described in other insects. Neuroanatomical analysis and two-photon calcium imaging demonstrate that DALcl1 and DALcl2 form two parallel channels, establishing connections with R neurons located in the peripheral and central domains of the EB, respectively. Although neurons of both lineages preferentially respond to bright objects, DALcl1 neurons have small ipsilateral, retinotopically ordered receptive fields, whereas DALcl2 neurons share a large excitatory receptive field in the contralateral hemifield. DALcl2 neurons become inhibited when the object enters the ipsilateral hemifield and display an additional excitation after the object leaves the field of view. Thus, the spatial position of a bright feature, such as a celestial body, may be encoded within this pathway.


Subject(s)
Cell Lineage , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Neurons/cytology , Neurons/physiology , Visual Pathways/physiology , Animals , Brain/cytology , Brain/physiology , Cells, Cultured , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/physiology , Neuropil/cytology , Neuropil/physiology , Visual Pathways/anatomy & histology
6.
J Comp Neurol ; 525(2): 363-379, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27350102

ABSTRACT

The Drosophila dopaminergic (DAergic) system consists of a relatively small number of neurons clustered throughout the brain and ventral nerve cord. Previous work shows that clusters of DA neurons innervate different brain compartments, which in part accounts for functional diversity of the DA system. We analyzed the association between DA neuron clusters and specific brain lineages, developmental and structural units of the Drosophila brain that provide a framework of connections that can be followed throughout development. The hatching larval brain contains six groups of primary DA neurons (born in the embryo), which we assign to six distinct lineages. We can show that all larval DA clusters persist into the adult brain. Some clusters increase in cell number during late larval stages, whereas others do not become DA positive until early pupa. Ablating neuroblasts with hydroxyurea (HU) prior to onset of larval proliferation (generates secondary neurons) confirms that these added DA clusters are primary neurons born in the embryo, rather than secondary neurons. A single cluster that becomes DA positive in the late pupa, PAM1/lineage DALcm1/2, forms part of a secondary lineage that can be ablated by larval HU application. By supplying lineage information for each DA cluster, our analysis promotes further developmental and functional analyses of this important system of neurons. J. Comp. Neurol. 525:363-379, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/cytology , Dopaminergic Neurons/cytology , Drosophila/embryology , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Cell Lineage , Imaging, Three-Dimensional , Immunohistochemistry , Microscopy, Confocal
7.
Curr Opin Insect Sci ; 18: 96-104, 2016 12.
Article in English | MEDLINE | ID: mdl-27939718

ABSTRACT

Glia of vertebrates and invertebrates alike represents a diverse population of cells in the nervous system, divided into numerous classes with different structural and functional characteristics. In insects, glia fall within three basic classes: surface, cell body, and neuropil glia. Due to the glial subclass-specific markers and genetic tools available in Drosophila, it is possible to establish the progenitor origin of these different populations and reconstruct their migration and differentiation during development. We review, and posit when appropriate, recently elucidated aspects of glial developmental dynamics. In particular, we focus on the relationships between mature glial subclasses of the larval nervous system (primary glia), born in the embryo, and glia of the adult (secondary glia), generated in the larva.


Subject(s)
Drosophila/cytology , Animals , Cell Lineage , Nervous System/cytology , Neurogenesis , Neuroglia/cytology
8.
Dev Neurobiol ; 76(4): 434-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26178322

ABSTRACT

The Drosophila brain consists of a relatively small number of invariant, genetically determined lineages which provide a model to study the relationship between gene function and neuronal architecture. In following this long-term goal, we reconstruct the morphology (projection pattern and connectivity) and gene expression patterns of brain lineages throughout development. In this article, we focus on the secondary phase of lineage morphogenesis, from the reactivation of neuroblast proliferation in the first larval instar to the time when proliferation ends and secondary axon tracts have fully extended in the late third larval instar. We have reconstructed the location and projection of secondary lineages at close (4 h) intervals and produced a detailed map in the form of confocal z-projections and digital three-dimensional models of all lineages at successive larval stages. Based on these reconstructions, we could compare the spatio-temporal pattern of axon formation and morphogenetic movements of different lineages in normal brain development. In addition to wild type, we reconstructed lineage morphology in two mutant conditions. (1) Expressing the construct UAS-p35 which rescues programmed cell death we could systematically determine which lineages normally lose hemilineages to apoptosis. (2) so-Gal4-driven expression of dominant-negative EGFR ablated the optic lobe, which allowed us to conclude that the global centrifugal movement normally affecting the cell bodies of lateral lineages in the late larva is causally related to the expansion of the optic lobe, and that the central pattern of axonal projections of these lineages is independent of the presence or absence of the optic lobe.


Subject(s)
Cell Lineage/physiology , Cell Movement/physiology , Drosophila/growth & development , Drosophila/physiology , Animals , Animals, Genetically Modified , Brain/anatomy & histology , Brain/growth & development , Brain/physiology , Cell Death/physiology , Drosophila/anatomy & histology , Drosophila Proteins/metabolism , Imaging, Three-Dimensional , Immunohistochemistry , Larva , Microscopy, Confocal , Microscopy, Electron , Neural Pathways/anatomy & histology , Neural Pathways/growth & development , Neural Pathways/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology
9.
Elife ; 42015 Aug 28.
Article in English | MEDLINE | ID: mdl-26317380

ABSTRACT

Genetic techniques have shed new light on the organization of the neurons in the ventral nervous system of the fruit fly.


Subject(s)
Central Nervous System/anatomy & histology , Drosophila/anatomy & histology , Drosophila/physiology , Neurons/physiology , Animals
10.
Dev Biol ; 406(1): 14-39, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26141956

ABSTRACT

Fixed lineages derived from unique, genetically specified neuroblasts form the anatomical building blocks of the Drosophila brain. Neurons belonging to the same lineage project their axons in a common tract, which is labeled by neuronal markers. In this paper, we present a detailed atlas of the lineage-associated tracts forming the brain of the early Drosophila larva, based on the use of global markers (anti-Neuroglian, anti-Neurotactin, inscuteable-Gal4>UAS-chRFP-Tub) and lineage-specific reporters. We describe 68 discrete fiber bundles that contain axons of one lineage or pairs/small sets of adjacent lineages. Bundles enter the neuropil at invariant locations, the lineage tract entry portals. Within the neuropil, these fiber bundles form larger fascicles that can be classified, by their main orientation, into longitudinal, transverse, and vertical (ascending/descending) fascicles. We present 3D digital models of lineage tract entry portals and neuropil fascicles, set into relationship to commonly used, easily recognizable reference structures such as the mushroom body, the antennal lobe, the optic lobe, and the Fasciclin II-positive fiber bundles that connect the brain and ventral nerve cord. Correspondences and differences between early larval tract anatomy and the previously described late larval and adult lineage patterns are highlighted. Our L1 neuro-anatomical atlas of lineages constitutes an essential step towards following morphologically defined lineages to the neuroblasts of the early embryo, which will ultimately make it possible to link the structure and connectivity of a lineage to the expression of genes in the particular neuroblast that gives rise to that lineage. Furthermore, the L1 atlas will be important for a host of ongoing work that attempts to reconstruct neuronal connectivity at the level of resolution of single neurons and their synapses.


Subject(s)
Brain/embryology , Brain/metabolism , Drosophila/embryology , Larva/metabolism , Animals , Axons/metabolism , Brain/anatomy & histology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/metabolism , Cell Lineage , Drosophila/anatomy & histology , Drosophila/metabolism , Drosophila Proteins/biosynthesis , Larva/anatomy & histology , Membrane Glycoproteins/biosynthesis , Neurons/metabolism , Neuropil/metabolism
11.
Dev Biol ; 402(1): 32-47, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25773365

ABSTRACT

The Drosophila brain is comprised of neurons formed by approximately 100 lineages, each of which is derived from a stereotyped, asymmetrically dividing neuroblast. Lineages serve as structural and developmental units of Drosophila brain anatomy and reconstruction of lineage projection patterns represents a suitable map of Drosophila brain circuitry at the level of neuron populations ("macro-circuitry"). Two phases of neuroblast proliferation, the first in the embryo and the second during the larval phase (following a period of mitotic quiescence), produce primary and secondary lineages, respectively. Using temporally controlled pulses of hydroxyurea (HU) to ablate neuroblasts and their corresponding secondary lineages during the larval phase, we analyzed the effect on development of primary and secondary lineages in the late larval and adult brain. Our findings indicate that timing of neuroblast re-activation is highly stereotyped, allowing us to establish "birth dates" for all secondary lineages. Furthermore, our results demonstrate that, whereas the trajectory and projection pattern of primary and secondary lineages is established in a largely independent manner, the final branching pattern of secondary neurons is dependent upon the presence of appropriate neuronal targets. Taken together, our data provide new insights into the degree of neuronal plasticity during Drosophila brain development.


Subject(s)
Brain/embryology , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neurons/metabolism , Animals , Cell Lineage , Cell Proliferation , Drosophila Proteins/metabolism , Hydroxyurea/chemistry , Microscopy, Confocal , Neurogenesis , Neurons/drug effects , Neurons/physiology , Time Factors
12.
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
13.
Dev Biol ; 384(2): 258-89, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23872236

ABSTRACT

The Drosophila central brain is largely composed of lineages, units of sibling neurons derived from a single progenitor cell or neuroblast. During the early embryonic period, neuroblasts generate the primary neurons that constitute the larval brain. Neuroblasts reactivate in the larva, adding to their lineages a large number of secondary neurons which, according to previous studies in which selected lineages were labeled by stably expressed markers, differentiate during metamorphosis, sending terminal axonal and dendritic branches into defined volumes of the brain neuropil. We call the overall projection pattern of neurons forming a given lineage the "projection envelope" of that lineage. By inducing MARCM clones at the early larval stage, we labeled the secondary progeny of each neuroblast. For the supraesophageal ganglion excluding mushroom body (the part of the brain investigated in the present work) we obtained 81 different types of clones. Based on the trajectory of their secondary axon tracts (described in the accompanying paper, Lovick et al., 2013), we assigned these clones to specific lineages defined in the larva. Since a labeled clone reveals all aspects (cell bodies, axon tracts, terminal arborization) of a lineage, we were able to describe projection envelopes for all secondary lineages of the supraesophageal ganglion. This work provides a framework by which the secondary neurons (forming the vast majority of adult brain neurons) can be assigned to genetically and developmentally defined groups. It also represents a step towards the goal to establish, for each lineage, the link between its mature anatomical and functional phenotype, and the genetic make-up of the neuroblast it descends from.


Subject(s)
Brain/growth & development , Drosophila/growth & development , Animals , Cell Lineage , Microscopy, Confocal
14.
Dev Biol ; 384(2): 228-57, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23880429

ABSTRACT

Neurons of the Drosophila central brain fall into approximately 100 paired groups, termed lineages. Each lineage is derived from a single asymmetrically-dividing neuroblast. Embryonic neuroblasts produce 1,500 primary neurons (per hemisphere) that make up the larval CNS followed by a second mitotic period in the larva that generates approximately 10,000 secondary, adult-specific neurons. Clonal analyses based on previous works using lineage-specific Gal4 drivers have established that such lineages form highly invariant morphological units. All neurons of a lineage project as one or a few axon tracts (secondary axon tracts, SATs) with characteristic trajectories, thereby representing unique hallmarks. In the neuropil, SATs assemble into larger fiber bundles (fascicles) which interconnect different neuropil compartments. We have analyzed the SATs and fascicles formed by lineages during larval, pupal, and adult stages using antibodies against membrane molecules (Neurotactin/Neuroglian) and synaptic proteins (Bruchpilot/N-Cadherin). The use of these markers allows one to identify fiber bundles of the adult brain and associate them with SATs and fascicles of the larval brain. This work lays the foundation for assigning the lineage identity of GFP-labeled MARCM clones on the basis of their close association with specific SATs and neuropil fascicles, as described in the accompanying paper (Wong et al., 2013. Postembryonic lineages of the Drosophila brain: II. Identification of lineage projection patterns based on MARCM clones. Submitted.).


Subject(s)
Body Patterning , Brain/growth & development , Drosophila/growth & development , Animals , Humans , Metamorphosis, Biological
15.
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
16.
J Comp Neurol ; 519(4): 661-89, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21246549

ABSTRACT

Most neurons of the central complex belong to 10 secondary (larvally produced) lineages. In the late larva, undifferentiated axon tracts of these lineages form a primordium in which all of the compartments of the central complex can be recognized as discrete entities. Four posterior lineages (DPMm1, DPMpm1, DPMpm2, and CM4) generate the classes of small-field neurons that interconnect the protocerebral bridge, fan-shaped body, noduli, and ellipsoid body. Three lineages located in the anterior brain, DALv2, BAmv1, and DALcl2, form the large-field neurons of the ellipsoid body and fan-shaped body, respectively. These lineages provide an input channel from the optic tubercle and connect the central complex with adjacent anterior brain compartments. Three lineages in the posterior cortex, CM3, CP2, and DPMpl2, connect the posterior brain neuropil with specific layers of the fan-shaped body. Even though all of the compartments of the central complex are prefigured in the late larval brain by the axon tracts of the above-mentioned lineages, the neuropil differentiates during the first 2 days of the pupal period when terminal branches and synapses of secondary neurons are formed. During this phase the initially straight horizontal layers of the central complex bend in the frontal plane, which produces the characteristic shape of the fan-shaped and ellipsoid body. Our analysis provides a comprehensive picture of the lineages that form the central complex, and will facilitate future studies that address the structure or function of the central complex at the single cell level.


Subject(s)
Cell Lineage , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/growth & development , Neurons/physiology , Animals , Brain/cytology , Brain/growth & development , Drosophila Proteins/metabolism , Metamorphosis, Biological , Morphogenesis , Neurons/cytology , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...