Abstract

Numerous neuropeptide systems have been implicated to coordinately control energy homeostasis, both centrally and peripherally. However, the vertebrate neuropeptide Y (NPY) system has emerged as the best described one regarding this biological process. The protostomian ortholog of NPY is neuropeptide F, characterized by an RXRF(Y)amide carboxyterminal motif. A second neuropeptide system is short NPF, characterized by an M/T/L/FRF(W)amide carboxyterminal motif. Although both short and long NPF neuropeptide systems display carboxyterminal sequence similarities, they are evolutionary distant and likely already arose as separate signaling systems in the common ancestor of deuterostomes and protostomes, indicating the functional importance of both. Both NPF and short-NPF systems seem to have roles in the coordination of feeding across bilaterian species, but during chordate evolution, the short NPF system appears to have been lost or evolved into the prolactin releasing peptide signaling system, which regulates feeding and has been suggested to be orthologous to sNPF. Here we review the roles of both NPF and sNPF systems in the regulation of feeding and metabolism in invertebrates.

Highlights

  • The nomenclature of the two distinct neuropeptide families, short neuropeptide F on the one hand, and neuropeptide F (NPF) on the other hand, has led to confusing data and conclusions in literature

  • We summarize work on the regulation of feeding and metabolism by NPF and short neuropeptide F (sNPF) in invertebrates and attempt to clarify annotation ambiguities that have become apparent upon large-scale phylogenomic analyses of NPF and sNPF systems across bilaterians [1, 2]

  • S. invicta, a downregulation of sNPF receptor (sNPFR) transcription was observed in mated queens that were starved, compared to well-fed congeners [31], again suggesting the importance of sNPF signaling during feeding or metabolically active states

Read more

Summary

Introduction

The nomenclature of the two distinct neuropeptide families, short neuropeptide F (sNPF) on the one hand, and neuropeptide F (NPF) on the other hand, has led to confusing data and conclusions in literature. The Drosophila NPF receptor can be activated by mammalian NPY-type neuropeptides when expressed in Xenopus oocytes [27]. Several studies in insects have demonstrated overlapping functions of NPF and sNPF signaling with respect to feeding and metabolism, suggesting a common evolutionary origin of sNPF and NPF neuropeptides and receptors.

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call