Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin

Publikation: Bidrag til tidsskriftTidsskriftartikelfagfællebedømt

Standard

Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin. / Zandawala, Meet; Yurgel, Maria E.; Texada, Michael J.; Liao, Sifang; Rewitz, Kim F.; Keene, Alex C.; Nässel, Dick R.

I: PLOS Genetics, Bind 14, Nr. 11, e1007767, 2018, s. 1-31.

Publikation: Bidrag til tidsskriftTidsskriftartikelfagfællebedømt

Harvard

Zandawala, M, Yurgel, ME, Texada, MJ, Liao, S, Rewitz, KF, Keene, AC & Nässel, DR 2018, 'Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin', PLOS Genetics, bind 14, nr. 11, e1007767, s. 1-31. https://doi.org/10.1371/journal.pgen.1007767

APA

Zandawala, M., Yurgel, M. E., Texada, M. J., Liao, S., Rewitz, K. F., Keene, A. C., & Nässel, D. R. (2018). Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin. PLOS Genetics, 14(11), 1-31. [e1007767]. https://doi.org/10.1371/journal.pgen.1007767

Vancouver

Zandawala M, Yurgel ME, Texada MJ, Liao S, Rewitz KF, Keene AC o.a. Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin. PLOS Genetics. 2018;14(11):1-31. e1007767. https://doi.org/10.1371/journal.pgen.1007767

Author

Zandawala, Meet ; Yurgel, Maria E. ; Texada, Michael J. ; Liao, Sifang ; Rewitz, Kim F. ; Keene, Alex C. ; Nässel, Dick R. / Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin. I: PLOS Genetics. 2018 ; Bind 14, Nr. 11. s. 1-31.

Bibtex

@article{2e7ee7ad074946ca98df405421a19028,
title = "Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin",
abstract = "Behavior and physiology are orchestrated by neuropeptides acting as central neuromodulators and circulating hormones. An outstanding question is how these neuropeptides function to coordinate complex and competing behaviors. In Drosophila, the neuropeptide leucokinin (LK) modulates diverse functions, but mechanisms underlying these complex interactions remain poorly understood. As a first step towards understanding these mechanisms, we delineated LK circuitry that governs various aspects of post-feeding physiology and behavior. We found that impaired LK signaling in Lk and Lk receptor (Lkr) mutants affects diverse but coordinated processes, including regulation of stress, water homeostasis, feeding, locomotor activity, and metabolic rate. Next, we sought to define the populations of LK neurons that contribute to the different aspects of this physiology. We find that the calcium activity in abdominal ganglia LK neurons (ABLKs), but not in the two sets of brain neurons, increases specifically following water consumption, suggesting that ABLKs regulate water homeostasis and its associated physiology. To identify targets of LK peptide, we mapped the distribution of Lkr expression, mined a brain single-cell transcriptome dataset for genes coexpressed with Lkr, and identified synaptic partners of LK neurons. Lkr expression in the brain insulin-producing cells (IPCs), gut, renal tubules and chemosensory cells, correlates well with regulatory roles detected in the Lk and Lkr mutants. Furthermore, these mutants and flies with targeted knockdown of Lkr in IPCs displayed altered expression of insulin-like peptides (DILPs) and transcripts in IPCs and increased starvation resistance. Thus, some effects of LK signaling appear to occur via DILP action. Collectively, our data suggest that the three sets of LK neurons have different targets, but modulate the establishment of post-prandial homeostasis by regulating distinct physiological processes and behaviors such as diuresis, metabolism, organismal activity and insulin signaling. These findings provide a platform for investigating feeding-related neuroendocrine regulation of vital behavior and physiology.",
author = "Meet Zandawala and Yurgel, {Maria E.} and Texada, {Michael J.} and Sifang Liao and Rewitz, {Kim F.} and Keene, {Alex C.} and N{\"a}ssel, {Dick R.}",
year = "2018",
doi = "10.1371/journal.pgen.1007767",
language = "English",
volume = "14",
pages = "1--31",
journal = "P L o S Genetics",
issn = "1553-7390",
publisher = "Public Library of Science",
number = "11",

}

RIS

TY - JOUR

T1 - Modulation of Drosophila post-feeding physiology and behavior by the neuropeptide leucokinin

AU - Zandawala, Meet

AU - Yurgel, Maria E.

AU - Texada, Michael J.

AU - Liao, Sifang

AU - Rewitz, Kim F.

AU - Keene, Alex C.

AU - Nässel, Dick R.

PY - 2018

Y1 - 2018

N2 - Behavior and physiology are orchestrated by neuropeptides acting as central neuromodulators and circulating hormones. An outstanding question is how these neuropeptides function to coordinate complex and competing behaviors. In Drosophila, the neuropeptide leucokinin (LK) modulates diverse functions, but mechanisms underlying these complex interactions remain poorly understood. As a first step towards understanding these mechanisms, we delineated LK circuitry that governs various aspects of post-feeding physiology and behavior. We found that impaired LK signaling in Lk and Lk receptor (Lkr) mutants affects diverse but coordinated processes, including regulation of stress, water homeostasis, feeding, locomotor activity, and metabolic rate. Next, we sought to define the populations of LK neurons that contribute to the different aspects of this physiology. We find that the calcium activity in abdominal ganglia LK neurons (ABLKs), but not in the two sets of brain neurons, increases specifically following water consumption, suggesting that ABLKs regulate water homeostasis and its associated physiology. To identify targets of LK peptide, we mapped the distribution of Lkr expression, mined a brain single-cell transcriptome dataset for genes coexpressed with Lkr, and identified synaptic partners of LK neurons. Lkr expression in the brain insulin-producing cells (IPCs), gut, renal tubules and chemosensory cells, correlates well with regulatory roles detected in the Lk and Lkr mutants. Furthermore, these mutants and flies with targeted knockdown of Lkr in IPCs displayed altered expression of insulin-like peptides (DILPs) and transcripts in IPCs and increased starvation resistance. Thus, some effects of LK signaling appear to occur via DILP action. Collectively, our data suggest that the three sets of LK neurons have different targets, but modulate the establishment of post-prandial homeostasis by regulating distinct physiological processes and behaviors such as diuresis, metabolism, organismal activity and insulin signaling. These findings provide a platform for investigating feeding-related neuroendocrine regulation of vital behavior and physiology.

AB - Behavior and physiology are orchestrated by neuropeptides acting as central neuromodulators and circulating hormones. An outstanding question is how these neuropeptides function to coordinate complex and competing behaviors. In Drosophila, the neuropeptide leucokinin (LK) modulates diverse functions, but mechanisms underlying these complex interactions remain poorly understood. As a first step towards understanding these mechanisms, we delineated LK circuitry that governs various aspects of post-feeding physiology and behavior. We found that impaired LK signaling in Lk and Lk receptor (Lkr) mutants affects diverse but coordinated processes, including regulation of stress, water homeostasis, feeding, locomotor activity, and metabolic rate. Next, we sought to define the populations of LK neurons that contribute to the different aspects of this physiology. We find that the calcium activity in abdominal ganglia LK neurons (ABLKs), but not in the two sets of brain neurons, increases specifically following water consumption, suggesting that ABLKs regulate water homeostasis and its associated physiology. To identify targets of LK peptide, we mapped the distribution of Lkr expression, mined a brain single-cell transcriptome dataset for genes coexpressed with Lkr, and identified synaptic partners of LK neurons. Lkr expression in the brain insulin-producing cells (IPCs), gut, renal tubules and chemosensory cells, correlates well with regulatory roles detected in the Lk and Lkr mutants. Furthermore, these mutants and flies with targeted knockdown of Lkr in IPCs displayed altered expression of insulin-like peptides (DILPs) and transcripts in IPCs and increased starvation resistance. Thus, some effects of LK signaling appear to occur via DILP action. Collectively, our data suggest that the three sets of LK neurons have different targets, but modulate the establishment of post-prandial homeostasis by regulating distinct physiological processes and behaviors such as diuresis, metabolism, organismal activity and insulin signaling. These findings provide a platform for investigating feeding-related neuroendocrine regulation of vital behavior and physiology.

UR - http://www.scopus.com/inward/record.url?scp=85056803906&partnerID=8YFLogxK

U2 - 10.1371/journal.pgen.1007767

DO - 10.1371/journal.pgen.1007767

M3 - Journal article

C2 - 30457986

AN - SCOPUS:85056803906

VL - 14

SP - 1

EP - 31

JO - P L o S Genetics

JF - P L o S Genetics

SN - 1553-7390

IS - 11

M1 - e1007767

ER -

ID: 209826455