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.

  • Review Article
  • Published:

Drosophila melanogaster embryonic haemocytes: masters of multitasking

Key Points

  • Drosophila melanogaster has three types of haemocyte: plasmatocytes, crystal cells and lamellocytes. In the embryo, 95% of all haemocytes are plasmatocytes.

  • D. melanogaster haemocyte development occurs in two waves: the first within the embryonic head mesoderm and the second within a specialized organ in the larva known as the lymph gland.

  • Many parallels exist between blood-cell development in flies and vertebrates, and many of the transcription factors that are required for haematopoiesis in D. melanogaster are homologues of genes that are central to vertebrate haematopoiesis.

  • Embryonic plasmatocytes are professional phagocytes and are responsible for the clearance of all apoptotic-cell debris in the embryo.

  • Plasmatocytes are highly motile cells. During development they migrate from their point of origin and disperse throughout the embryo and will also rapidly migrate towards epithelial wounds in an inflammatory-like response.

  • Embryonic plasmatocytes are a main source of extracellular matrix proteins and are required for the normal development of some tissues, such as the central nervous system and developing gut.

Abstract

Drosophila melanogaster haemocytes constitute the cellular arm of a robust innate immune system in flies. In the adult and larva, these cells operate as the first line of defence against invading microorganisms: they phagocytose pathogens and produce antimicrobial peptides. However, in the sterile environment of the embryo, these important immune functions are largely redundant. Instead, throughout development, embryonic haemocytes are occupied with other tasks: they undergo complex migrations and carry out several non-immune functions that are crucial for successful embryogenesis.

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: Haemocyte development in Drosophila melanogaster.
Figure 2: Transcriptional regulation of haematopoiesis.
Figure 3: Developmental migrations.
Figure 4: Haemocyte migration mechanisms.
Figure 5: Multiple haemocyte stimuli.

Similar content being viewed by others

References

  1. Ribeiro, C. & Brehelin, M. Insect haemocytes: what type of cell is that? J. Insect Physiol. 52, 417–429 (2006).

    CAS  PubMed  Google Scholar 

  2. Lavine, M. D. & Strand, M. R. Insect hemocytes and their role in immunity. Insect Biochem. Mol. Biol. 32, 1295–1309 (2002).

    CAS  PubMed  Google Scholar 

  3. Holz, A., Bossinger, B., Strasser, T., Janning, W. & Klapper, R. The two origins of hemocytes in Drosophila. Development 130, 4955–4962 (2003). Demonstrates that embryonic haemocytes and lymph-gland haemocytes arise from two separate anlagen in the embryo.

    CAS  PubMed  Google Scholar 

  4. Tepass, U., Fessler, L. I., Aziz, A. & Hartenstein, V. Embryonic origin of hemocytes and their relationship to cell death in Drosophila. Development 120, 1829–1837 (1994). This paper was the first to identify the origin of embryonic plasmatocytes and their subsequent migration routes through the embryo.

    CAS  PubMed  Google Scholar 

  5. Lebestky, T., Chang, T., Hartenstein, V. & Banerjee, U. Specification of Drosophila hematopoietic lineage by conserved transcription factors. Science 288, 146–149 (2000).

    CAS  PubMed  Google Scholar 

  6. Bataille, L., Auge, B., Ferjoux, G., Haenlin, M. & Waltzer, L. Resolving embryonic blood cell fate choice in Drosophila: interplay of GCM and RUNX factors. Development 132, 4635–4644 (2005). Shows that GCM and GCM2 antagonize crystal-cell development and that plasmatocytes and crystal cells develop from the same bipolar progenitors in the embryo.

    CAS  PubMed  Google Scholar 

  7. Mandal, L., Banerjee, U. & Hartenstein, V. Evidence for a fruit fly hemangioblast and similarities between lymph-gland hematopoiesis in fruit fly and mammal aorta-gonadal-mesonephros mesoderm. Nature Genet. 36, 1019–1023 (2004). This important paper presents evidence for the existence of a haemangioblast cell in Drosophila melanogaster embryos that can give rise to two daughter cells: one that differentiates into heart or aorta and another that differentiates into blood.

    CAS  PubMed  Google Scholar 

  8. Lanot, R., Zachary, D., Holder, F. & Meister, M. Postembryonic hematopoiesis in Drosophila. Dev. Biol. 230, 243–257 (2001).

    CAS  PubMed  Google Scholar 

  9. Zettervall, C. J., et al. A directed screen for genes involved in Drosophila blood cell activation. Proc. Natl Acad. Sci. USA 101, 14192–14197 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jung, S. H., Evans, C. J., Uemura, C. & Banerjee, U. The Drosophila lymph gland as a developmental model of hematopoiesis. Development 132, 2521–2533 (2005). Characterizes lymph-gland development in great detail by showing the existence of distinct zones of haemocyte maturation, signalling and proliferation within the developing lymph gland.

    CAS  PubMed  Google Scholar 

  11. Rehorn, K. P., Thelen, H., Michelson, A. M. & Reuter, R. A molecular aspect of hematopoiesis and endoderm development common to vertebrates and Drosophila. Development 122, 4023–4031 (1996).

    CAS  PubMed  Google Scholar 

  12. Waltzer, L., Bataille, L., Peyrefitte, S. & Haenlin, M. Two isoforms of Serpent containing either one or two GATA zinc fingers have different roles in Drosophila haematopoiesis. EMBO J. 21, 5477–5486 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Alfonso, T. B. & Jones, B. W. gcm2 promotes glial cell differentiation and is required with glial cells missing for macrophage development in Drosophila. Dev. Biol. 248, 369–383 (2002).

    CAS  PubMed  Google Scholar 

  14. Bernardoni, R., Vivancos, V. & Giangrande, A. glide/gcm is expressed and required in the scavenger cell lineage. Dev. Biol. 191, 118–130 (1997).

    CAS  PubMed  Google Scholar 

  15. de Bruijn, M. F. & Speck, N. A. Core-binding factors in hematopoiesis and immune function. Oncogene 23, 4238–4248 (2004).

    CAS  PubMed  Google Scholar 

  16. Waltzer, L., Ferjoux, G., Bataille, L. & Haenlin, M. Cooperation between the GATA and RUNX factors Serpent and Lozenge during Drosophila hematopoiesis. EMBO J. 22, 6516–6525 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Fossett, N., et al. The Friend of GATA proteins U-shaped, FOG-1, and FOG-2 function as negative regulators of blood, heart, and eye development in Drosophila. Proc. Natl Acad. Sci. USA 98, 7342–7347 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lebestky, T., Jung, S. H. & Banerjee, U. A Serrate-expressing signaling center controls Drosophila hematopoiesis. Genes Dev. 17, 348–353 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Crozatier, M., Ubeda, J. M., Vincent, A. & Meister, M. Cellular immune response to parasitization in Drosophila requires the EBF orthologue collier. PLoS Biol 2, e196 (2004).

    PubMed  PubMed Central  Google Scholar 

  20. Munier, A. I., et al. PVF2, a PDGF/VEGF-like growth factor, induces hemocyte proliferation in Drosophila larvae. EMBO Rep. 3, 1195–1200 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Bruckner, K., et al. The PDGF/VEGF receptor controls blood cell survival in Drosophila. Dev. Cell 7, 73–84 (2004). Shows the requirement of the PDGF–VEGF receptor (PVR) for the survival of embryonic haemocytes in Drosophila melanogaster.

    PubMed  Google Scholar 

  22. Heino, T. I., et al. The Drosophila VEGF receptor homolog is expressed in hemocytes. Mech. Dev. 109, 69–77 (2001).

    CAS  PubMed  Google Scholar 

  23. Harrison, D. A., Binari, R., Nahreini, T. S., Gilman, M. & Perrimon, N. Activation of a Drosophila Janus kinase (JAK) causes hematopoietic neoplasia and developmental defects. EMBO J. 14, 2857–2865 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Hou, S. X., Zheng, Z., Chen, X. & Perrimon, N. The Jak/STAT pathway in model organisms: emerging roles in cell movement. Dev. Cell 3, 765–778 (2002).

    CAS  PubMed  Google Scholar 

  25. Luo, H., Hanratty, W. P. & Dearolf, C. R. An amino acid substitution in the Drosophila hop Tum-l Jak kinase causes leukemia-like hematopoietic defects. EMBO J. 14, 1412–1420 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Remillieux-Leschelle, N., Santamaria, P. & Randsholt, N. B. Regulation of larval hematopoiesis in Drosophila melanogaster: a role for the multi sex combs gene. Genetics 162, 1259–1274 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Sorrentino, R. P., Melk, J. P. & Govind, S. Genetic analysis of contributions of dorsal group and JAK-Stat92E pathway genes to larval hemocyte concentration and the egg encapsulation response in Drosophila. Genetics 166, 1343–1356 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Lemaitre, B. The road to Toll. Nature Rev. Immunol. 4, 521–527 (2004).

    CAS  Google Scholar 

  29. Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J. M. & Hoffmann, J. A. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell 86, 973–983 (1996).

    CAS  PubMed  Google Scholar 

  30. Hultmark, D. Drosophila immunity: paths and patterns. Curr. Opin. Immunol. 15, 12–19 (2003).

    CAS  PubMed  Google Scholar 

  31. Leclerc, V. & Reichhart, J. M. The immune response of Drosophila melanogaster. Immunol. Rev. 198, 59–71 (2004).

    CAS  PubMed  Google Scholar 

  32. Qiu, P., Pan, P. C. & Govind, S. A role for the Drosophila Toll/Cactus pathway in larval hematopoiesis. Development 125, 1909–1920 (1998).

    CAS  PubMed  Google Scholar 

  33. Gerttula, S., Jin, Y. S. & Anderson, K. V. Zygotic expression and activity of the Drosophila Toll gene, a gene required maternally for embryonic dorsal–ventral pattern formation. Genetics 119, 123–133 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Govind, S. Rel signalling pathway and the melanotic tumour phenotype of Drosophila. Biochem. Soc. Trans. 24, 39–44 (1996).

    CAS  PubMed  Google Scholar 

  35. Matova, N. & Anderson, K. V. Rel/NF-κB double mutants reveal that cellular immunity is central to Drosophila host defense. Proc. Natl Acad. Sci. USA 103, 16424–16429 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Agaisse, H., Petersen, U. M., Boutros, M., Mathey-Prevot, B. & Perrimon, N. Signaling role of hemocytes in Drosophila JAK/STAT-dependent response to septic injury. Dev. Cell 5, 441–450 (2003).

    CAS  PubMed  Google Scholar 

  37. Pevny, L., et al. Erythroid differentiation in chimaeric mice blocked by a targeted mutation in the gene for transcription factor GATA-1. Nature 349, 257–260 (1991).

    CAS  PubMed  Google Scholar 

  38. Tsai, F. Y., et al. An early haematopoietic defect in mice lacking the transcription factor GATA-2. Nature 371, 221–226 (1994).

    CAS  PubMed  Google Scholar 

  39. Ting, C. N., Olson, M. C., Barton, K. P. & Leiden, J. M. Transcription factor GATA-3 is required for development of the T-cell lineage. Nature 384, 474–478 (1996).

    CAS  PubMed  Google Scholar 

  40. Elagib, K. E., et al. RUNX1 and GATA-1 coexpression and cooperation in megakaryocytic differentiation. Blood 101, 4333–4341 (2003).

    CAS  PubMed  Google Scholar 

  41. Tsang, A. P., et al. FOG, a multitype zinc finger protein, acts as a cofactor for transcription factor GATA-1 in erythroid and megakaryocytic differentiation. Cell 90, 109–119 (1997).

    CAS  PubMed  Google Scholar 

  42. Tsang, A. P., Fujiwara, Y., Hom, D. B. & Orkin, S. H. Failure of megakaryopoiesis and arrested erythropoiesis in mice lacking the GATA-1 transcriptional cofactor FOG. Genes Dev. 12, 1176–1188 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Crispino, J. D., Lodish, M. B., MacKay, J. P. & Orkin, S. H. Use of altered specificity mutants to probe a specific protein–protein interaction in differentiation: the GATA-1:FOG complex. Mol. Cell 3, 219–228 (1999).

    CAS  PubMed  Google Scholar 

  44. Allman, D., Aster, J. C. & Pear, W. S. Notch signaling in hematopoiesis and early lymphocyte development. Immunol. Rev. 187, 75–86 (2002).

    CAS  PubMed  Google Scholar 

  45. Ohishi, K., Katayama, N., Shiku, H., Varnum-Finney, B. & Bernstein, I. D. Notch signalling in hematopoiesis. Semin. Cell Dev. Biol. 14, 143–150 (2003).

    CAS  PubMed  Google Scholar 

  46. Gerber, H. P., et al. VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature 417, 954–958 (2002).

    CAS  PubMed  Google Scholar 

  47. Rane, S. G. & Reddy, E. P. JAKs, STATs and Src kinases in hematopoiesis. Oncogene 21, 3334–3358 (2002).

    CAS  PubMed  Google Scholar 

  48. Bromberg, J. Stat proteins and oncogenesis. J. Clin. Invest. 109, 1139–1142 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Rayet, B. & Gelinas, C. Aberrant rel/nfkb genes and activity in human cancer. Oncogene 18, 6938–6947 (1999).

    CAS  PubMed  Google Scholar 

  50. Hagman, J., Belanger, C., Travis, A., Turck, C. W. & Grosschedl, R. Cloning and functional characterization of early B-cell factor, a regulator of lymphocyte-specific gene expression. Genes Dev. 7, 760–773 (1993).

    CAS  PubMed  Google Scholar 

  51. Crozatier, M., Valle, D., Dubois, L., Ibnsouda, S. & Vincent, A. Collier, a novel regulator of Drosophila head development, is expressed in a single mitotic domain. Curr. Biol. 6, 707–718 (1996).

    CAS  PubMed  Google Scholar 

  52. Lin, H. & Grosschedl, R. Failure of B-cell differentiation in mice lacking the transcription factor EBF. Nature 376, 263–267 (1995).

    CAS  PubMed  Google Scholar 

  53. Maier, H. & Hagman, J. Roles of EBF and Pax-5 in B lineage commitment and development. Semin. Immunol. 14, 415–422 (2002).

    CAS  PubMed  Google Scholar 

  54. Evans, C. J., Hartenstein, V. & Banerjee, U. Thicker than blood: conserved mechanisms in Drosophila and vertebrate hematopoiesis. Dev. Cell 5, 673–690 (2003). This is an excellent detailed review that covers Drosophila melanogaster blood-cell development and its parallels with vertebrate haematopoiesis.

    CAS  PubMed  Google Scholar 

  55. Hartenstein, V. Blood cells and blood cell development in the animal kingdom. Annu. Rev. Cell Dev. Biol. 22, 677–712 (2006).

    CAS  PubMed  Google Scholar 

  56. Wood, W., et al. Mesenchymal cells engulf and clear apoptotic footplate cells in macrophageless PU.1 null mouse embryos. Development 127, 5245–5252 (2000).

    CAS  PubMed  Google Scholar 

  57. Sonnenfeld, M. J. & Jacobs, J. R. Macrophages and glia participate in the removal of apoptotic neurons from the Drosophila embryonic nervous system. J. Comp. Neurol. 359, 644–652 (1995).

    CAS  PubMed  Google Scholar 

  58. Diez-Roux, G. & Lang, R. A. Macrophages induce apoptosis in normal cells in vivo. Development 124, 3633–3638 (1997).

    CAS  PubMed  Google Scholar 

  59. Franc, N. C., Dimarcq, J. L., Lagueux, M., Hoffmann, J. & Ezekowitz, R. A. Croquemort, a novel Drosophila hemocyte/macrophage receptor that recognizes apoptotic cells. Immunity 4, 431–443 (1996).

    CAS  PubMed  Google Scholar 

  60. Franc, N. C., Heitzler, P., Ezekowitz, R. A. & White, K. Requirement for croquemort in phagocytosis of apoptotic cells in Drosophila. Science 284, 1991–1994 (1999).

    CAS  PubMed  Google Scholar 

  61. Fadok, V. A., et al. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature 405, 85–90 (2000).

    CAS  PubMed  Google Scholar 

  62. Henson, P. M., Bratton, D. L. & Fadok, V. A. Apoptotic cell removal. Curr. Biol. 11, R795–R805 (2001).

    CAS  PubMed  Google Scholar 

  63. Freeman, M. R., Delrow, J., Kim, J., Johnson, E. & Doe, C. Q. Unwrapping glial biology: gcm target genes regulating glial development, diversification, and function. Neuron 38, 567–580 (2003).

    CAS  PubMed  Google Scholar 

  64. Manaka, J., et al. Draper-mediated and phosphatidylserine-independent phagocytosis of apoptotic cells by Drosophila hemocytes/macrophages. J. Biol. Chem. 279, 48466–48476 (2004).

    CAS  PubMed  Google Scholar 

  65. Mergliano, J. & Minden, J. S. Caspase-independent cell engulfment mirrors cell death pattern in Drosophila embryos. Development 130, 5779–5789 (2003).

    CAS  PubMed  Google Scholar 

  66. Wood, W., Faria, C. & Jacinto, A. Distinct mechanisms regulate hemocyte chemotaxis during development and wound healing in Drosophila melanogaster. J. Cell Biol. 173, 405–416 (2006). Shows that embryonic haemocytes use two different mechanisms to undergo directional migration towards different stimuli.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Cho, N. K., et al. Developmental control of blood cell migration by the Drosophila VEGF pathway. Cell 108, 865–876 (2002).

    CAS  PubMed  Google Scholar 

  68. Stramer, B., et al. Live imaging of wound inflammation in Drosophila embryos reveals key roles for small GTPases during in vivo cell migration. J. Cell Biol. 168, 567–573 (2005). Characterizes the inflammatory-like chemotaxis of embryonic Drosophila melanogaster haemocytes towards epithelial wounds and the role of the Rho-family small GTPases in this process.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Paladi, M. & Tepass, U. Function of Rho GTPases in embryonic blood cell migration in Drosophila. J. Cell Sci. 117, 6313–6326 (2004).

    CAS  PubMed  Google Scholar 

  70. Huelsmann, S., Hepper, C., Marchese, D., Knoll, C. & Reuter, R. The PDZ-GEF dizzy regulates cell shape of migrating macrophages via Rap1 and integrins in the Drosophila embryo. Development 133, 2915–2924 (2006).

    CAS  PubMed  Google Scholar 

  71. Worthylake, R. A., Lemoine, S., Watson, J. M. & Burridge, K. RhoA is required for monocyte tail retraction during transendothelial migration. J. Cell Biol. 154, 147–160 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Etienne-Manneville, S. Cdc42 — the centre of polarity. J. Cell Sci. 117, 1291–1300 (2004).

    CAS  PubMed  Google Scholar 

  73. Stephens, L., Ellson, C. & Hawkins, P. Roles of PI3Ks in leukocyte chemotaxis and phagocytosis. Curr. Opin. Cell Biol. 14, 203–213 (2002).

    CAS  PubMed  Google Scholar 

  74. Weiner, O. D. Regulation of cell polarity during eukaryotic chemotaxis: the chemotactic compass. Curr. Opin. Cell Biol. 14, 196–202 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Williams, M. J., Wiklund, M. L., Wikman, S. & Hultmark, D. Rac1 signalling in the Drosophila larval cellular immune response. J. Cell Sci. 119, 2015–2024 (2006).

    CAS  PubMed  Google Scholar 

  76. Fessler, J. H. & Fessler, L. I. Drosophila extracellular matrix. Annu. Rev. Cell Biol. 5, 309–339 (1989).

    CAS  PubMed  Google Scholar 

  77. Fogerty, F. J., et al. Tiggrin, a novel Drosophila extracellular matrix protein that functions as a ligand for Drosophila αPS2 βPS integrins. Development 120, 1747–1758 (1994).

    CAS  PubMed  Google Scholar 

  78. Nelson, R. E., et al. Peroxidasin: a novel enzyme-matrix protein of Drosophila development. EMBO J. 13, 3438–3447 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Kramerova, I. A., et al. Papilin in development; a pericellular protein with a homology to the ADAMTS metalloproteinases. Development 127, 5475–5485 (2000).

    CAS  PubMed  Google Scholar 

  80. Martinek, N., Zou, R., Berg, M., Sodek, J. & Ringuette, M. Evolutionary conservation and association of SPARC with the basal lamina in Drosophila. Dev. Genes Evol. 212, 124–133 (2002).

    CAS  PubMed  Google Scholar 

  81. Hortsch, M., et al. The expression of MDP-1, a component of Drosophila embryonic basement membranes, is modulated by apoptotic cell death. Int. J. Dev. Biol. 42, 33–42 (1998).

    CAS  PubMed  Google Scholar 

  82. Kusche-Gullberg, M., Garrison, K., MacKrell, A. J., Fessler, L. I. & Fessler, J. H. Laminin A chain: expression during Drosophila development and genomic sequence. EMBO J. 11, 4519–4527 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Mirre, C., Cecchini, J. P., Le Parco, Y. & Knibiehler, B. De novo expression of a type IV collagen gene in Drosophila embryos is restricted to mesodermal derivatives and occurs at germ band shortening. Development 102, 369–376 (1988).

    CAS  PubMed  Google Scholar 

  84. Knibiehler, B., Mirre, C., Cecchini, J. P. & Le Parco, Y. Haemocytes accumulate collagen transcripts during Drosophila melanogaster metamorphosis. Roux's Arch. Dev. Biol. 196, 243–247 (1987).

    Google Scholar 

  85. Le Parco, Y., Le Bivic, A., Knibiehler, B., Mirre, C. & Cecchini, J. P. DCg1 αIV collagen chain of Drosophila melanogaster is synthesized during embryonic organogenesis by mesenchymal cells and is deposited in muscle basement membranes. Insect Biochem. 19, 789–802 (1989).

    CAS  Google Scholar 

  86. Yasothornsrikul, S., Davis, W. J., Cramer, G., Kimbrell, D. A. & Dearolf, C. R. viking: identification and characterization of a second type IV collagen in Drosophila. Gene 198, 17–25 (1997).

    CAS  PubMed  Google Scholar 

  87. Olofsson, B. & Page, D. T. Condensation of the central nervous system in embryonic Drosophila is inhibited by blocking hemocyte migration or neural activity. Dev. Biol. 279, 233–243 (2005).

    CAS  PubMed  Google Scholar 

  88. Brown, N. H. Null mutations in the aPS2 and bPS integrin subunit genes have distinct phenotypes. Development 120, 1221–1231 (1994).

    CAS  PubMed  Google Scholar 

  89. Yarnitzky, T. & Volk, T. Laminin is required for heart, somatic muscles, and gut development in the Drosophila embryo. Dev. Biol. 169, 609–618 (1995).

    CAS  PubMed  Google Scholar 

  90. Sears, H. C., Kennedy, C. J. & Garrity, P. A. Macrophage-mediated corpse engulfment is required for normal Drosophila CNS morphogenesis. Development 130, 3557–3565 (2003).

    CAS  PubMed  Google Scholar 

  91. Abrams, J. M., White, K., Fessler, L. I. & Steller, H. Programmed cell death during Drosophila embryogenesis. Development 117, 29–43 (1993).

    CAS  PubMed  Google Scholar 

  92. Zhou, L., Hashimi, H., Schwartz, L. M. & Nambu, J. R. Programmed cell death in the Drosophila central nervous system midline. Curr. Biol. 5, 784–790 (1995).

    CAS  PubMed  Google Scholar 

  93. Lauber, K., et al. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 113, 717–730 (2003).

    CAS  PubMed  Google Scholar 

  94. Rizki, T. M. in Physiology of Insect Development (ed. Campbell, F. L.) 91–94 (Chicago University Press, Illinois, 1956).

    Google Scholar 

  95. Rizki, T. M. & Rizki, R. M. Properties of the larval hemocytes of Drosophila melanogaster. Experientia 36, 1223–1226 (1980).

    Google Scholar 

  96. Hoffmann, J. A. & Reichhart, J. M. Drosophila innate immunity: an evolutionary perspective. Nature Immunol. 3, 121–126 (2002).

    CAS  Google Scholar 

  97. Rizki, T. M. Alterations in the hemocyte population of Drosophila melanogaster. J. Morphol. 100, 437–458 (1957).

    Google Scholar 

  98. Shrestha, R. & Gateff, E. Ultrastructure and cytochemistry of the cell types in the larval hematopoietic organs and hemolymph of Drosophila melanogaster. Develop. Growth and Differ. 24, 65–82 (1982).

    Google Scholar 

  99. Rizki, T. M. & Rizki, R. M. in Insect Ultrastructure (ed. King, R. C.) 579–604 (Plenum, New York, 1984).

    Google Scholar 

  100. Rizki, T. M. & Rizki, R. M. Lamellocyte differentiation in Drosophila larvae parasitized by Leptopilina. Dev. Comp. Immunol. 16, 103–110 (1992).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank B. Stramer for helpful feedback on the manuscript. W.W. is funded by the Wellcome Trust and A.J. is funded by Fundação para a Ciência e a Tecnologia, Instituto Gulbenkian de Ciência and by the Network of Excellence Cells into Organs, supported by the European Union Framework Programme 6.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Will Wood's homepage

Glossary

Mesoderm

A morphologically distinct cell layer that can be recognized in the early embryos of most bilaterian phyla. It gives rise to tissues that are interposed between ectodermal and endodermal epithelia, including muscle, connective and blood tissue.

Anlage

An initial clustering of embryonic cells that serves as a foundation from which a body part or an organ develops.

Proventriculus

A structure that is located at the caudal end of the oesophagus, formed at the junction of the foregut and the midgut. It serves as a valve that regulates the passage of food into the midgut.

Melanization

A reaction that is used as an immune mechanism in arthropods to encapsulate and kill microbial pathogens. Arthropod melanization is controlled by a cascade of serine proteases that ultimately activates the enzyme prophenoloxidase, which, in turn, catalyses the synthesis of melanin.

Unipotent

A cell that has the capacity to differentiate into only one type of tissue or cell.

Oligopotent

A cell that has the capacity to give rise to several cell types.

Haemangioblast

A multipotent cell that is a common precursor to haematopoietic and endothelial cells. In Drosophila melanogaster, haemangioblasts can give rise to haemocytes or to heart and aorta cells.

Haemocoel

A cavity or series of spaces between the organs of organisms with open circulatory systems.

Gram-positive bacteria

Gram staining is an empirical method of differentiating bacterial species into two groups based on structural differences within their cell walls. Gram-positive bacteria are those that retain the dark-blue dye crystal violet, whereas Gram-negative bacteria do not.

Phagocyte

A cell that ingests and destroys cell debris or foreign matter, such as microorganisms, through a process known as phagocytosis.

Phosphatidylserine receptor

The receptor for phosphatidylserine. It is expressed in phagocytic cells and is used for the detection of apoptotic cells.

Filopodium

A thin rod-like structure that extends from the cell membrane. It contains long actin filaments that are crosslinked into bundles by actin-binding proteins.

Lamellipodium

A two-dimensional actin meshwork that extends from the leading edge of many migrating cells.

Cofilin

An actin-binding protein that causes depolymerization at the minus end of actin filaments.

Haemolymph

A combination of lymph and interstitial fluid that circulates through the haemocoel.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wood, W., Jacinto, A. Drosophila melanogaster embryonic haemocytes: masters of multitasking. Nat Rev Mol Cell Biol 8, 542–551 (2007). https://doi.org/10.1038/nrm2202

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2202

This article is cited by

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