Cargando…

High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo

In multicellular organisms, insulin/IGF signaling (IIS) plays a central role in matching energy needs with uptake and storage, participating in functions as diverse as metabolic homeostasis, growth, reproduction and ageing. In mammals, this pleiotropy of action relies in part on a dichotomy of actio...

Descripción completa

Detalles Bibliográficos
Autores principales: Pasco, Matthieu Y., Léopold, Pierre
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3342234/
https://www.ncbi.nlm.nih.gov/pubmed/22567167
http://dx.doi.org/10.1371/journal.pone.0036583
_version_ 1782231660972998656
author Pasco, Matthieu Y.
Léopold, Pierre
author_facet Pasco, Matthieu Y.
Léopold, Pierre
author_sort Pasco, Matthieu Y.
collection PubMed
description In multicellular organisms, insulin/IGF signaling (IIS) plays a central role in matching energy needs with uptake and storage, participating in functions as diverse as metabolic homeostasis, growth, reproduction and ageing. In mammals, this pleiotropy of action relies in part on a dichotomy of action of insulin, IGF-I and their respective membrane-bound receptors. In organisms with simpler IIS, this functional separation is questionable. In Drosophila IIS consists of several insulin-like peptides called Dilps, activating a unique membrane receptor and its downstream signaling cascade. During larval development, IIS is involved in metabolic homeostasis and growth. We have used feeding conditions (high sugar diet, HSD) that induce an important change in metabolic homeostasis to monitor possible effects on growth. Unexpectedly we observed that HSD-fed animals exhibited severe growth inhibition as a consequence of peripheral Dilp resistance. Dilp-resistant animals present several metabolic disorders similar to those observed in type II diabetes (T2D) patients. By exploring the molecular mechanisms involved in Drosophila Dilp resistance, we found a major role for the lipocalin Neural Lazarillo (NLaz), a target of JNK signaling. NLaz expression is strongly increased upon HSD and animals heterozygous for an NLaz null mutation are fully protected from HSD-induced Dilp resistance. NLaz is a secreted protein homologous to the Retinol-Binding Protein 4 involved in the onset of T2D in human and mice. These results indicate that insulin resistance shares common molecular mechanisms in flies and human and that Drosophila could emerge as a powerful genetic system to study some aspects of this complex syndrome.
format Online
Article
Text
id pubmed-3342234
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-33422342012-05-07 High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo Pasco, Matthieu Y. Léopold, Pierre PLoS One Research Article In multicellular organisms, insulin/IGF signaling (IIS) plays a central role in matching energy needs with uptake and storage, participating in functions as diverse as metabolic homeostasis, growth, reproduction and ageing. In mammals, this pleiotropy of action relies in part on a dichotomy of action of insulin, IGF-I and their respective membrane-bound receptors. In organisms with simpler IIS, this functional separation is questionable. In Drosophila IIS consists of several insulin-like peptides called Dilps, activating a unique membrane receptor and its downstream signaling cascade. During larval development, IIS is involved in metabolic homeostasis and growth. We have used feeding conditions (high sugar diet, HSD) that induce an important change in metabolic homeostasis to monitor possible effects on growth. Unexpectedly we observed that HSD-fed animals exhibited severe growth inhibition as a consequence of peripheral Dilp resistance. Dilp-resistant animals present several metabolic disorders similar to those observed in type II diabetes (T2D) patients. By exploring the molecular mechanisms involved in Drosophila Dilp resistance, we found a major role for the lipocalin Neural Lazarillo (NLaz), a target of JNK signaling. NLaz expression is strongly increased upon HSD and animals heterozygous for an NLaz null mutation are fully protected from HSD-induced Dilp resistance. NLaz is a secreted protein homologous to the Retinol-Binding Protein 4 involved in the onset of T2D in human and mice. These results indicate that insulin resistance shares common molecular mechanisms in flies and human and that Drosophila could emerge as a powerful genetic system to study some aspects of this complex syndrome. Public Library of Science 2012-05-02 /pmc/articles/PMC3342234/ /pubmed/22567167 http://dx.doi.org/10.1371/journal.pone.0036583 Text en Pasco, Léopold. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Pasco, Matthieu Y.
Léopold, Pierre
High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title_full High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title_fullStr High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title_full_unstemmed High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title_short High Sugar-Induced Insulin Resistance in Drosophila Relies on the Lipocalin Neural Lazarillo
title_sort high sugar-induced insulin resistance in drosophila relies on the lipocalin neural lazarillo
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3342234/
https://www.ncbi.nlm.nih.gov/pubmed/22567167
http://dx.doi.org/10.1371/journal.pone.0036583
work_keys_str_mv AT pascomatthieuy highsugarinducedinsulinresistanceindrosophilareliesonthelipocalinneurallazarillo
AT leopoldpierre highsugarinducedinsulinresistanceindrosophilareliesonthelipocalinneurallazarillo