Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Integration of temporal and spatial patterning generates neural diversity

Abstract

In the Drosophila optic lobes, 800 retinotopically organized columns in the medulla act as functional units for processing visual information. The medulla contains over 80 types of neuron, which belong to two classes: uni-columnar neurons have a stoichiometry of one per column, while multi-columnar neurons contact multiple columns. Here we show that combinatorial inputs from temporal and spatial axes generate this neuronal diversity: all neuroblasts switch fates over time to produce different neurons; the neuroepithelium that generates neuroblasts is also subdivided into six compartments by the expression of specific factors. Uni-columnar neurons are produced in all spatial compartments independently of spatial input; they innervate the neuropil where they are generated. Multi-columnar neurons are generated in smaller numbers in restricted compartments and require spatial input; the majority of their cell bodies subsequently move to cover the entire medulla. The selective integration of spatial inputs by a fixed temporal neuroblast cascade thus acts as a powerful mechanism for generating neural diversity, regulating stoichiometry and the formation of retinotopy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The OPC neuroepithelium is patterned along its dorsal–ventral axis.
Figure 2: Distinct neuronal cell types are generated along the dorsal–ventral axis of the OPC.
Figure 3: Temporal and spatial inputs are required for neuronal specification in the medulla.
Figure 4: Spatial genes cross-regulate each other in the OPC neuroepithelium.
Figure 5: Neuronal movement during medulla development is restricted to multi-columnar cell types.

Similar content being viewed by others

References

  1. Fischbach, K. F. D. & Dittrich, A. P. The optic lobe of Drosophila melanogaster. I. A Golgi analysis of wild-type structure. Cell Tissue Res . 258, 441–475 (1989)

    Article  Google Scholar 

  2. Morante, J. & Desplan, C. The color-vision circuit in the medulla of Drosophila. Curr. Biol. 18, 553–565 (2008)

    Article  CAS  Google Scholar 

  3. Bausenwein, B., Dittrich, A. P. & Fischbach, K. F. The optic lobe of Drosophila melanogaster. II. Sorting of retinotopic pathways in the medulla. Cell Tissue Res . 267, 17–28 (1992)

    Article  CAS  Google Scholar 

  4. Gao, S. et al. The neural substrate of spectral preference in Drosophila. Neuron 60, 328–342 (2008)

    Article  CAS  Google Scholar 

  5. Rister, J. et al. Dissection of the peripheral motion channel in the visual system of Drosophila melanogaster. Neuron 56, 155–170 (2007)

    Article  CAS  Google Scholar 

  6. Nassif, C., Noveen, A. & Hartenstein, V. Early development of the Drosophila brain: III. The pattern of neuropile founder tracts during the larval period. J. Comp. Neurol. 455, 417–434 (2003)

    Article  Google Scholar 

  7. Egger, B., Boone, J. Q., Stevens, N. R., Brand, A. H. & Doe, C. Q. Regulation of spindle orientation and neural stem cell fate in the Drosophila optic lobe. Neural Dev. 2, 1 (2007)

    Article  Google Scholar 

  8. Huang, Z. & Kunes, S. Signals transmitted along retinal axons in Drosophila: Hedgehog signal reception and the cell circuitry of lamina cartridge assembly. Development 125, 3753–3764 (1998)

    CAS  PubMed  Google Scholar 

  9. Egger, B., Gold, K. S. & Brand, A. H. Notch regulates the switch from symmetric to asymmetric neural stem cell division in the Drosophila optic lobe. Development 137, 2981–2987 (2010)

    Article  CAS  Google Scholar 

  10. Yasugi, T., Umetsu, D., Murakami, S., Sato, M. & Tabata, T. Drosophila optic lobe neuroblasts triggered by a wave of proneural gene expression that is negatively regulated by JAK/STAT. Development 135, 1471–1480 (2008)

    Article  CAS  Google Scholar 

  11. Yasugi, T., Sugie, A., Umetsu, D. & Tabata, T. Coordinated sequential action of EGFR and Notch signaling pathways regulates proneural wave progression in the Drosophila optic lobe. Development 137, 3193–3203 (2010)

    Article  CAS  Google Scholar 

  12. Ngo, K. T. et al. Concomitant requirement for Notch and Jak/Stat signaling during neuro-epithelial differentiation in the Drosophila optic lobe. Dev. Biol. 346, 284–295 (2010)

    Article  CAS  Google Scholar 

  13. Li, X. et al. Temporal patterning of Drosophila medulla neuroblasts controls neural fates. Nature 498, 456–462 (2013)

    Article  ADS  CAS  Google Scholar 

  14. Suzuki, T., Kaido, M., Takayama, R. & Sato, M. A temporal mechanism that produces neuronal diversity in the Drosophila visual center. Dev. Biol. 380, 12–24 (2013)

    Article  CAS  Google Scholar 

  15. Isshiki, T., Pearson, B., Holbrook, S. & Doe, C. Q. Drosophila neuroblasts sequentially express transcription factors which specify the temporal identity of their neuronal progeny. Cell 106, 511–521 (2001)

    Article  CAS  Google Scholar 

  16. Brody, T. & Odenwald, W. F. Programmed transformations in neuroblast gene expression during Drosophila CNS lineage development. Dev. Biol. 226, 34–44 (2000)

    Article  CAS  Google Scholar 

  17. Grosskortenhaus, R., Pearson, B. J., Marusich, A. & Doe, C. Q. Regulation of temporal identity transitions in Drosophila neuroblasts. Dev. Cell 8, 193–202 (2005)

    Article  CAS  Google Scholar 

  18. Erclik, T., Hartenstein, V., Lipshitz, H. D. & McInnes, R. R. Conserved role of the Vsx genes supports a monophyletic origin for bilaterian visual systems. Curr. Biol. 18, 1278–1287 (2008)

    Article  CAS  Google Scholar 

  19. Gold, K. S. & Brand, A. H. Optix defines a neuroepithelial compartment in the optic lobe of the Drosophila brain. Neural Dev. 9, 18 (2014)

    Article  Google Scholar 

  20. Chang, T., Mazotta, J., Dumstrei, K., Dumitrescu, A. & Hartenstein, V. Dpp and Hh signaling in the Drosophila embryonic eye field. Development 128, 4691–4704 (2001)

    CAS  PubMed  Google Scholar 

  21. Kaphingst, K. & Kunes, S. Pattern formation in the visual centers of the Drosophila brain: wingless acts via decapentaplegic to specify the dorsoventral axis. Cell 78, 437–448 (1994)

    Article  CAS  Google Scholar 

  22. Bertet, C. et al. Temporal patterning of neuroblasts controls Notch-mediated cell survival through regulation of Hid or Reaper. Cell 158, 1173–1186 (2014)

    Article  CAS  Google Scholar 

  23. Chen, Z. et al. A unique class of neural progenitors in the Drosophila optic lobe generates both migrating neurons and glia. Cell Rep. 15, 774–786 (2016)

    Article  CAS  Google Scholar 

  24. Hasegawa, E. et al. Concentric zones, cell migration and neuronal circuits in the Drosophila visual center. Development 138, 983–993 (2011)

    Article  CAS  Google Scholar 

  25. Skeath, J. B., Zhang, Y., Holmgren, R., Carroll, S. B. & Doe, C. Q. Specification of neuroblast identity in the Drosophila embryonic central nervous system by gooseberry-distal. Nature 376, 427–430 (1995)

    Article  ADS  CAS  Google Scholar 

  26. McDonald, J. A. et al. Dorsoventral patterning in the Drosophila central nervous system: the vnd homeobox gene specifies ventral column identity. Genes Dev. 12, 3603–3612 (1998)

    Article  CAS  Google Scholar 

  27. Technau, G. M., Berger, C. & Urbach, R. Generation of cell diversity and segmental pattern in the embryonic central nervous system of Drosophila. Dev. Dyn. 235, 861–869 (2006)

    Article  CAS  Google Scholar 

  28. Karlsson D., Baumgardt M. & Thor S. Segment-specific neuronal subtype specification by the integration of anteroposterior and temporal cues. http://dx.doi.org/10.1371/journal.pbio.1000368 (2010)

  29. Morante, J. & Desplan, C. Dissection and staining of Drosophila optic lobes at different stages of development. Cold Spring Harb. Protocols 2011, 652–656 (2011)

    Google Scholar 

Download references

Acknowledgements

We thank the fly community for gifts of antibodies and fly stocks, D. Vasiliauskas and R. Johnston for collaborating on screening the modENCODE antibodies, and the Desplan laboratory members for discussion and support. This work was supported by a grant from the National Institutes of Health (NIH) to C.D. (R01 EY017916), to T.E. by the Canadian Institutes of Health Research (CIHR) and the Natural Sciences and Engineering Research Council of Canada (NSERC: RGPIN-2015-06457) to X.L. by The Robert Leet and Clara Guthrie Patterson Trust Postdoctoral Fellowship and to C.B. by fellowships from EMBO (ALTF 680-2009) and HFSPO (LT000077/2010-L). The modENCODE antibodies were produced with support of NIH grant U01HG004264 awarded to K.P.W.

Author information

Authors and Affiliations

Authors

Contributions

C.D., T.E., X.L. and M.C. planned the project and analysed the data; X.L., T.E. and C.B. performed the antibody screen; T.E. and M.C. conducted experiments with the regional and temporal genes, and analysed cellular movements; X.L. focused on the Tsh/Pm1/Pm2 and 27b-Gal4 experiments. R.B. and J.N. contributed to the analysis of Rx mutants. U.A. contributed to the analysis of cOPC axonal projections and C.K. helped M.C. with experiments. Z.C. performed the Optix>G-Trace and Vsx1-Gal4 single neuron flip-out experiments. R.B. generated Tm2-lexA flies. L.S., N.N. and K.P.W. generated the modENCODE antibodies that were used in the screen. The manuscript was written by T.E., M.C. and C.D., and commented on by X.L. and C.B.

Corresponding author

Correspondence to Claude Desplan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks C. Doe and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Compartmentalization of the OPC.

a, Cross-section of the OPC stained for Dac in red (lamina), E-cad in blue (NE) and Deadpan-lacZ in green (neuroblasts and neurons). The NE is converted into medulla neuroblasts on its medial side and lamina precursors on its lateral side. b, Early second-instar larval brain. Rx (blue), Optix-Gal4 driving UAS-nuGFP (green) and Vsx1 (red) label distinct regions of the OPC. The two Optix>nuGFP cells located between the pOPC arms are outside the OPC neuroepithelium. c, Surface view of the third-instar larval OPC. Vsx1 (red) marks the cOPC and the majority of cOPC neurons. Optix (green) labels the mOPC (both dorsal and ventral) and E-cadherin (blue) outlines the OPC neuroepithelium. d, Regionalization of the optic anlage in the stage 13 embryo, labelled by FasII (blue): Rx (green) and Vsx1 (red) are expressed in the pOPC and cOPC primordia, respectively.

Extended Data Figure 2 Regionalization of the Hth neuronal crescent.

a, Surface view of the third-instar larval OPC. In the cOPC, Lim3 (blue) is expressed in the ap-negative (ap-lacZ in green) population of Hth neurons (red) and Ey neurons. b, Cross-section view of the third-instar larval OPC. In the pOPC, Lim3 (blue) is expressed in the ap-negative (ap-lacZ in green) population of Hth neurons (red). c, Lateral view of the third-instar larval OPC. The 27b>GFP neurons (green) include larger Hth-positive (red) neurons in the pOPC, and smaller Hth-negative neurons (Tm1) born in a later temporal stage throughout the OPC. d, Lateral view of the third-instar larval OPC. The 27b>GFP neurons (green) are Svp-positive (blue) in the pOPC and Tsh-positive (red) in the ventral pOPC. e, Pm3 neuronal clone in the adult labelled by MARCM with the hth-Gal4 driver. Vsx1 is marked in red and Svp in blue. f, Neurons in the adult brain marked by flip out clones with the 27b-Gal4 driver (GFP in green). One Pm1 and one Pm2 neuron are labelled in the medulla rim. The cell bodies for both neurons express Hth (red and arrowheads in bottom panel). Note that a GFP-positive cell body in the medulla cortex (Tm1 neuron) does not express Hth (arrow in bottom panel). The medulla neuropil is labelled with N-cadherin (blue).

Extended Data Figure 3 Fate of the NOFF neurons in different mutant conditions.

af, Lateral view of larval optic lobe neurons with Lim3 in red and Hth in blue. a, In WT cOPC, NOFF Lim3+Hth+ neurons are intermingled with Lim3-Hth+ neurons (the Mi1 Bsh+ neurons); whereas in the mOPC, Hth+ neurons are not intermingled with NOFF Lim3+ neurons. b, c, In Dronc mutants, cell death of Lim3+ NOFF neurons is prevented and Hth+ cells are intermingled with Lim3+ neurons. These Lim3+ neurons are not Svp+ (c). d, Apoptosis is selectively abolished in mOPC neurons by overexpressing P35 in all Optix-derived neurons (marked by GFP, in green). Hth+ cells are intermingled with Lim3+ neurons. e, In Vsx1 RNAi clones marked by GFP in green, there are no Hth+Lim3+neurons. f, g, Rx LOF mutation leads to loss of Hth+Lim3+neurons in the pOPC. h, Overexpression of Optix in the cOPC leads to loss of Hth+Lim3+ neurons.

Extended Data Figure 4 Temporal and spatial genes control cell-type specification in the OPC.

a, The hth mutant MARCM clone in the pOPC. Tsh expression (red) is lost in the clone. b, Wild-type third-instar larval brain. Lateral view of the Hth neuronal crescent (green). Svp (blue) is co-expressed with Hth in the cOPC and dorsal and ventral pOPC. Svp-negative neurons express Bsh (red). Arrows point to the Svp cells that are generated by the wingless-positive half of the pOPC (tOPC). c, Rx mutant third-instar larval brain. Lateral view of the Hth neuronal crescent (green). Svp expression (blue) is maintained in the cOPC but lost in the pOPC. Asterisks denote where the pOPC is located. Bsh expression (red) is unaffected. Arrows point to the Svp cells that are generated by the wingless-positive half of the pOPC (tOPC). d, Lateral view of the third-instar medulla cortex. Svp (green) is ectopically expressed in Rx GOF clones. The Pm1 marker Tsh (red) is ectopically expressed in ventral mOPC clones (solid arrow) but is not in clones in the dorsal mOPC or ventral cOPC (outlined arrows). e, f, Lateral view of third-instar larval brains stained for Tsh (green) and Svp (blue). e, In wild-type brains, Svp is expressed in the cOPC, tOPC and dorsal and ventral pOPC clusters. Tsh is co-expressed with Svp in ventral OPC neurons. f, In Rx mutant brains, Svp expression remains in the cOPC and tOPC but is lost in the pOPC. Tsh expression is also lost in the ventral OPC. g, Wild-type adult optic lobe. Hth (green) and Svp (blue) are co-expressed in populations of neurons in the dorsal and ventral medulla rim. h, Rx mutant adult optic lobe. The Hth (green) and Svp (blue) co-expressing populations are greatly reduced in number or absent in the medulla rim. i, Wild-type adult optic lobe with 27b-Gal4 driving GFP expression (green) in Pm1 and Pm2 neurons in the medulla rim and Tm1 neurons in the cortex. The neuropil is labelled in red with N-cadherin. j, In Rx mutant adult optic lobe, Pm1 and Pm2 neurons are absent from the medulla rim (boxed region). Tm1 neurons are unaffected. k, Optix mutant MARCM clone in the ventral mOPC. Svp (cyan) is derepressed in the clone (arrow in k) but neither Vsx1 (red in k) or Tsh (magenta in bottom panel of k) is activated.

Extended Data Figure 5 cOPC-derived neurons move during pupal stages.

ac, Vsx1>>lacZ lineage trace analysis (β-gal in red) in the medulla cortex at P0 (a), P20 (b) and P30 (c). cOPC-derived neurons remained clustered until P20. At P30 they have moved to cover the entire dorsal–ventral axis of the medulla cortex. Arrow denotes distal enrichment of cells in the central region of the cortex. d, Vsx1>>lacZ lineage trace analysis (β-gal in red) in the adult optic lobe. cOPC-derived neurons are located throughout the dorsal–ventral axis of the medulla cortex, and their cell bodies are enriched proximally with the exception of the central cortex, which also contains a distal enrichment of cell bodies. Vsx1 (green) is enriched proximally.

Extended Data Figure 6 Hth neurons during pupal development.

ad, Developing medulla cortex and rim at P10 (a), P20 (b, c) and P80 (d). Bsh neurons (red) remain evenly distributed across the dorsal–ventral axis of the medulla cortex at all stages. In contrast, Pm3 neurons marked by the co-expression of Hth (green) and Svp (blue) in the cOPC remain clustered until P20. Pm1 and Pm2 neurons are also clustered at P10 and P20 (arrows in c). e, Adult optic lobe stained for Bsh (red). Bsh neurons are found throughout the dorsal–ventral axis of the cortex, and their cell bodies are located in the distal cortex.

Extended Data Figure 7 Movement of Pm3 neurons in the pupa.

a, Surface view of the medulla cortex at P20. The neurons born in the cOPC, marked by Vsx1 in red, form a stripe that transverses the cortex. Hth neurons form a band of cells that covers the cortex in the dorsal–ventral axis. Pm3 neurons are marked by the triple-label of Vsx1, Hth (green) and Svp (blue), and they remain clustered in the Vsx1 stripe. b, Surface view of the medulla cortex at P40. The Pm3 neurons have moved throughout the dorsal–ventral axis of the cortex. Vsx1 neurons are no longer clustered together. c, Optic lobe at P50. Vsx1 and Pm3 neurons are found throughout the medulla cortex. Pm1 and Pm2 neurons have not moved, and remain clustered at the posterior regions, which were previously the OPC and are now the developing medulla rim. d, Adult optic lobe. Vsx1 (red) and Hth (green) neurons are dispersed throughout the dorsal–ventral axis of the medulla cortex. Pm3 neurons (marked by the overlap of Hth and Vsx1) and Vsx1 neurons in general have cell bodies located in the proximal half of the cortex.

Extended Data Figure 8 Uni- and multi-columnar neurons in the adult medulla.

a, Lateral view of the third-instar larval brain. Tm1 neurons (the smaller cells labelled with 27b-Gal4 driving UAS-GFP in green) are born throughout the OPC. The cOPC is labelled with Vsx1 (red). b, Adult optic lobe. The cell bodies of Tm1 neurons are located in the distal medulla cortex along the entire dorsal–ventral axis. The medulla neuropil is marked by N-cadherin in magenta. c, The hh lineage-trace in the adult medulla. Ventrally derived cells (β-gal in blue) are located throughout the medulla cortex but only overlap with Bsh (red) and Tm2-LexA>GFP (Tm2 neurons, green) in the ventral half (magnified in the right-hand panel). Note that Mi1 and Tm2 nuclei are located in the distal cortex. Dashed line denotes the dorsal–ventral boundary. d, R83H09-Gal4 driving CD8::GFP in TmY14 neurons represents an example of a medulla neuronal type produced by the cOPC that does not migrate. Their cell bodies can be found in the middle of the medulla cortex whereas their projections span the entire medulla neuropil. e, MARCM clone generated in the third-instar larva and visualized in the adult medulla (green). N-cadherin labels the neuropil in magenta. Cells in clones labelled by a Gal4 driver for the uni-columnar cell type Mi1 remain clustered in the ventral half of the medulla.

Extended Data Figure 9 Vsx-positive cell types in the medulla.

Thirteen Vsx-positive neuron types found in the adult medulla. Neurons are labelled with Vsx-Gal4 driving the expression of flip-out-GFP after heat-shock induction. The name of the neuron is included, unless the morphology could not be unambiguously assigned to a published cell type (e, l and m). All Vsx-positive neurons identified are multi-columnar in morphology.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Erclik, T., Li, X., Courgeon, M. et al. Integration of temporal and spatial patterning generates neural diversity. Nature 541, 365–370 (2017). https://doi.org/10.1038/nature20794

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature20794

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing