Cargando…

Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism

BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outco...

Descripción completa

Detalles Bibliográficos
Autores principales: Anderson, Jacqueline M., Boardman, Amber A., Bates, Rhiannon, Zou, Xunchang, Huang, Wei, Cao, Lei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9997972/
https://www.ncbi.nlm.nih.gov/pubmed/36893171
http://dx.doi.org/10.1371/journal.pone.0282566
_version_ 1784903372515573760
author Anderson, Jacqueline M.
Boardman, Amber A.
Bates, Rhiannon
Zou, Xunchang
Huang, Wei
Cao, Lei
author_facet Anderson, Jacqueline M.
Boardman, Amber A.
Bates, Rhiannon
Zou, Xunchang
Huang, Wei
Cao, Lei
author_sort Anderson, Jacqueline M.
collection PubMed
description BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outcomes. Brain-derived neurotrophic factor (Bdnf) and its receptor tropomyosin kinase receptor B (Ntrk2) were upregulated in the hypothalamus, hippocampus, and amygdala by implementing EE in BTBR mice, suggesting that BDNF-TrkB signaling plays a role in the EE-BTBR phenotype. Here, we used an adeno-associated virus (AAV) vector to overexpress the TrkB full-length (TrkB.FL) BDNF receptor in the BTBR mouse hypothalamus in order to assess whether hypothalamic BDNF-TrkB signaling is responsible for the improved metabolic and behavioral phenotypes associated with EE. Normal chow diet (NCD)-fed and high fat diet (HFD)-fed BTBR mice were randomized to receive either bilateral injections of AAV-TrkB.FL or AAV-YFP as control, and were subjected to metabolic and behavioral assessments up to 24 weeks post-injection. Both NCD and HFD TrkB.FL overexpressing mice displayed improved metabolic outcomes, characterized as reduced percent weight gain and increased energy expenditure. NCD TrkB.FL mice showed improved glycemic control, reduced adiposity, and increased lean mass. In NCD mice, TrkB.FL overexpression altered the ratio of TrkB.FL/TrkB.T1 protein expression and increased phosphorylation of PLCγ in the hypothalamus. TrkB.FL overexpression also upregulated expression of hypothalamic genes involved in energy regulation and altered expression of genes involved in thermogenesis, lipolysis, and energy expenditure in white adipose tissue and brown adipose tissue. In HFD mice, TrkB.FL overexpression increased phosphorylation of PLCγ. TrkB.FL overexpression in the hypothalamus did not improve behavioral deficits in either NCD or HFD mice. Together, these results suggest that enhancing hypothalamic TrkB.FL signaling improves metabolic health in BTBR mice.
format Online
Article
Text
id pubmed-9997972
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-99979722023-03-10 Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism Anderson, Jacqueline M. Boardman, Amber A. Bates, Rhiannon Zou, Xunchang Huang, Wei Cao, Lei PLoS One Research Article BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outcomes. Brain-derived neurotrophic factor (Bdnf) and its receptor tropomyosin kinase receptor B (Ntrk2) were upregulated in the hypothalamus, hippocampus, and amygdala by implementing EE in BTBR mice, suggesting that BDNF-TrkB signaling plays a role in the EE-BTBR phenotype. Here, we used an adeno-associated virus (AAV) vector to overexpress the TrkB full-length (TrkB.FL) BDNF receptor in the BTBR mouse hypothalamus in order to assess whether hypothalamic BDNF-TrkB signaling is responsible for the improved metabolic and behavioral phenotypes associated with EE. Normal chow diet (NCD)-fed and high fat diet (HFD)-fed BTBR mice were randomized to receive either bilateral injections of AAV-TrkB.FL or AAV-YFP as control, and were subjected to metabolic and behavioral assessments up to 24 weeks post-injection. Both NCD and HFD TrkB.FL overexpressing mice displayed improved metabolic outcomes, characterized as reduced percent weight gain and increased energy expenditure. NCD TrkB.FL mice showed improved glycemic control, reduced adiposity, and increased lean mass. In NCD mice, TrkB.FL overexpression altered the ratio of TrkB.FL/TrkB.T1 protein expression and increased phosphorylation of PLCγ in the hypothalamus. TrkB.FL overexpression also upregulated expression of hypothalamic genes involved in energy regulation and altered expression of genes involved in thermogenesis, lipolysis, and energy expenditure in white adipose tissue and brown adipose tissue. In HFD mice, TrkB.FL overexpression increased phosphorylation of PLCγ. TrkB.FL overexpression in the hypothalamus did not improve behavioral deficits in either NCD or HFD mice. Together, these results suggest that enhancing hypothalamic TrkB.FL signaling improves metabolic health in BTBR mice. Public Library of Science 2023-03-09 /pmc/articles/PMC9997972/ /pubmed/36893171 http://dx.doi.org/10.1371/journal.pone.0282566 Text en © 2023 Anderson et al https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Anderson, Jacqueline M.
Boardman, Amber A.
Bates, Rhiannon
Zou, Xunchang
Huang, Wei
Cao, Lei
Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title_full Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title_fullStr Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title_full_unstemmed Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title_short Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism
title_sort hypothalamic trkb.fl overexpression improves metabolic outcomes in the btbr mouse model of autism
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9997972/
https://www.ncbi.nlm.nih.gov/pubmed/36893171
http://dx.doi.org/10.1371/journal.pone.0282566
work_keys_str_mv AT andersonjacquelinem hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism
AT boardmanambera hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism
AT batesrhiannon hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism
AT zouxunchang hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism
AT huangwei hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism
AT caolei hypothalamictrkbfloverexpressionimprovesmetabolicoutcomesinthebtbrmousemodelofautism