Cargando…

Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice

OBJECTIVE: This study aims to investigate whether hypoxia-inducible factor 1α (HIF1α) in the neurons of the mediobasal hypothalamus is involved in the regulation of body weight, glucose, and lipid metabolism in mice and to explore the underlying molecular mechanisms. METHODS: HIF1α (flox/flox) mice...

Descripción completa

Detalles Bibliográficos
Autores principales: Rozjan, Azmat, Shan, Weibi, Yao, Qiaoling
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8558296/
https://www.ncbi.nlm.nih.gov/pubmed/34733235
http://dx.doi.org/10.3389/fendo.2021.668193
_version_ 1784592526126088192
author Rozjan, Azmat
Shan, Weibi
Yao, Qiaoling
author_facet Rozjan, Azmat
Shan, Weibi
Yao, Qiaoling
author_sort Rozjan, Azmat
collection PubMed
description OBJECTIVE: This study aims to investigate whether hypoxia-inducible factor 1α (HIF1α) in the neurons of the mediobasal hypothalamus is involved in the regulation of body weight, glucose, and lipid metabolism in mice and to explore the underlying molecular mechanisms. METHODS: HIF1α (flox/flox) mice were used. The adeno-associated virus that contained either cre, GFP and syn, or GFP and syn (controls) was injected into the mediobasal hypothalamus to selectively knock out HIF1α in the neurons of the mediobasal hypothalamus. The body weight and food intake were weighed daily. The levels of blood glucose, insulin, total cholesterol (TC), triglyceride (TG), free fatty acid (FFA), high-density lipoprotein (HDL), and low-density lipoprotein (LDL)were tested. Intraperitoneal glucose tolerance test (IPGTT) was performed. The insulin-stimulated Akt phosphorylation in the liver, epididymal fat, and skeletal muscle were examined. Also, the mRNA expression levels of HIF1α, proopiomelanocortin (POMC), neuropeptide Y (NPY), and glucose transporter protein 4 (Glut4) in the hypothalamus were checked. RESULTS: After selectively knocking out HIF1α in the neurons of the mediobasal hypothalamus (HIF1αKOMBH), the body weights and food intake of mice increased significantly compared with the control mice (p < 0.001 at 4 weeks). Compared with that of the control group, the insulin level of HIF1αKOMBH mice was 3.5 times higher (p < 0.01). The results of the IPGTT showed that the blood glucose level of the HIF1αKOMBH group at 20–120 min was significantly higher than that of the control group (p < 0.05). The serum TC, FFA, HDL, and LDL content of the HIF1αKOMBH group was significantly higher than those of the control group (p < 0.05). Western blot results showed that compared with those in the control group, insulin-induced AKT phosphorylation levels in liver, epididymal fat, and skeletal muscle in the HIF1αKOMBH group were not as significantly elevated as in the control group. Reverse transcription-polymerase chain reaction (RT-PCR) results in the whole hypothalamus showed a significant decrease in Glut4 mRNA expression. And the mRNA expression levels of HIF1α, POMC, and NPY of the HIF1αKOMBH group decreased significantly in ventral hypothalamus. CONCLUSIONS: The hypothalamic neuronal HIF1α plays an important role in the regulation of body weight balance in mice under normoxic condition. In the absence of hypothalamic neuronal HIF1α, the mice gained weight with increased appetite, accompanied with abnormal glucose and lipid metabolism. POMC and Glut4 may be responsible for this effect of HIF1α.
format Online
Article
Text
id pubmed-8558296
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-85582962021-11-02 Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice Rozjan, Azmat Shan, Weibi Yao, Qiaoling Front Endocrinol (Lausanne) Endocrinology OBJECTIVE: This study aims to investigate whether hypoxia-inducible factor 1α (HIF1α) in the neurons of the mediobasal hypothalamus is involved in the regulation of body weight, glucose, and lipid metabolism in mice and to explore the underlying molecular mechanisms. METHODS: HIF1α (flox/flox) mice were used. The adeno-associated virus that contained either cre, GFP and syn, or GFP and syn (controls) was injected into the mediobasal hypothalamus to selectively knock out HIF1α in the neurons of the mediobasal hypothalamus. The body weight and food intake were weighed daily. The levels of blood glucose, insulin, total cholesterol (TC), triglyceride (TG), free fatty acid (FFA), high-density lipoprotein (HDL), and low-density lipoprotein (LDL)were tested. Intraperitoneal glucose tolerance test (IPGTT) was performed. The insulin-stimulated Akt phosphorylation in the liver, epididymal fat, and skeletal muscle were examined. Also, the mRNA expression levels of HIF1α, proopiomelanocortin (POMC), neuropeptide Y (NPY), and glucose transporter protein 4 (Glut4) in the hypothalamus were checked. RESULTS: After selectively knocking out HIF1α in the neurons of the mediobasal hypothalamus (HIF1αKOMBH), the body weights and food intake of mice increased significantly compared with the control mice (p < 0.001 at 4 weeks). Compared with that of the control group, the insulin level of HIF1αKOMBH mice was 3.5 times higher (p < 0.01). The results of the IPGTT showed that the blood glucose level of the HIF1αKOMBH group at 20–120 min was significantly higher than that of the control group (p < 0.05). The serum TC, FFA, HDL, and LDL content of the HIF1αKOMBH group was significantly higher than those of the control group (p < 0.05). Western blot results showed that compared with those in the control group, insulin-induced AKT phosphorylation levels in liver, epididymal fat, and skeletal muscle in the HIF1αKOMBH group were not as significantly elevated as in the control group. Reverse transcription-polymerase chain reaction (RT-PCR) results in the whole hypothalamus showed a significant decrease in Glut4 mRNA expression. And the mRNA expression levels of HIF1α, POMC, and NPY of the HIF1αKOMBH group decreased significantly in ventral hypothalamus. CONCLUSIONS: The hypothalamic neuronal HIF1α plays an important role in the regulation of body weight balance in mice under normoxic condition. In the absence of hypothalamic neuronal HIF1α, the mice gained weight with increased appetite, accompanied with abnormal glucose and lipid metabolism. POMC and Glut4 may be responsible for this effect of HIF1α. Frontiers Media S.A. 2021-10-18 /pmc/articles/PMC8558296/ /pubmed/34733235 http://dx.doi.org/10.3389/fendo.2021.668193 Text en Copyright © 2021 Rozjan, Shan and Yao https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Endocrinology
Rozjan, Azmat
Shan, Weibi
Yao, Qiaoling
Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title_full Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title_fullStr Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title_full_unstemmed Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title_short Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice
title_sort metabolic consequences of neuronal hif1α-deficiency in mediobasal hypothalamus in mice
topic Endocrinology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8558296/
https://www.ncbi.nlm.nih.gov/pubmed/34733235
http://dx.doi.org/10.3389/fendo.2021.668193
work_keys_str_mv AT rozjanazmat metabolicconsequencesofneuronalhif1adeficiencyinmediobasalhypothalamusinmice
AT shanweibi metabolicconsequencesofneuronalhif1adeficiencyinmediobasalhypothalamusinmice
AT yaoqiaoling metabolicconsequencesofneuronalhif1adeficiencyinmediobasalhypothalamusinmice