Cargando…

Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice

BACKGROUND: End-stage renal disease is primarily caused by diabetic kidney disease (DKD). The Farnesoid X receptor (FXR), a member of the nuclear receptor superfamily, has anti-inflammatory, lipid-lowering and hypoglycemic properties. It also inhibits renal fibrosis. Although its physiological role...

Descripción completa

Detalles Bibliográficos
Autores principales: Qiu, Yuxiang, Kang, Ningsu, Wang, Xi, Yao, Yao, Cui, Jun, Zhang, Xiaoyan, Zheng, Lu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: PeerJ Inc. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10544308/
https://www.ncbi.nlm.nih.gov/pubmed/37790634
http://dx.doi.org/10.7717/peerj.16155
_version_ 1785114477727842304
author Qiu, Yuxiang
Kang, Ningsu
Wang, Xi
Yao, Yao
Cui, Jun
Zhang, Xiaoyan
Zheng, Lu
author_facet Qiu, Yuxiang
Kang, Ningsu
Wang, Xi
Yao, Yao
Cui, Jun
Zhang, Xiaoyan
Zheng, Lu
author_sort Qiu, Yuxiang
collection PubMed
description BACKGROUND: End-stage renal disease is primarily caused by diabetic kidney disease (DKD). The Farnesoid X receptor (FXR), a member of the nuclear receptor superfamily, has anti-inflammatory, lipid-lowering and hypoglycemic properties. It also inhibits renal fibrosis. Although its physiological role is not fully understood, it also plays a role in the control of diabetic nephropathy (DN). METHODS: In the present study, we examined male FXR & leptin receptor double knockout mice, in which weight, blood glucose, body fat, and other indicators were monitored. After 6 months of rearing, blood and urine samples were collected and biochemical parameters were measured. Fibrosis was assessed by Masson’s stain, while the assessment of the resuscitation case’s condition was performed using succinate dehydrogenase (SDHA) stain immunohistochemistry, which measures aerobic respiration. Expression of molecules such as connective tissue growth factor (CTGF), SMAD family members 3 (Smad3) and 7 (Smad7), and small heterodimer partner were detected by RT-PCR and Western blotting as part of the application. RESULTS: FXR knockout decreased body weight and body fat in db/db mice, but increased blood glucose, urine output, and renal fibrosis. Primary mesangial cells (P-MCs) from FXR(+/ +) mice stimulated with transforming growth factor β1 (TGFβ1) showed significantly higher levels of related fibrosis factors, TGFβ1 and Smad3 mRNA and protein, and significantly reduced levels of Smad7. These effects were reversed by the action of FXR agonist chenodeoxycholic acid (CDCA). P-MCs from FXR(−/ −) mice stimulated with TGFβ1 resulted in an increase in the expression and protein levels of collagen I and TGFβ1, and the addition of CDCA had no significant effect on TGFβ1 stimulation. However, compared with FXR(+/ +)db/db mice, the rate of oxygen consumption, the rate of carbon dioxide production, and the rate of energy conversion were increased in FXR(−/ −)db/db mice, whereas the SDHA succinate dehydrogenase, a marker enzyme for aerobic respiration, was significantly decreased. CONCLUSIONS: These results provide evidence that FXR plays a critical role in the regulation of mesangial cells in DN. The likely mechanism is that aberrant FXR expression activates TGFβ1, which induces extracellular matrix accumulation through the classical Smad signaling pathway, leading to mitochondrial dysfunction.
format Online
Article
Text
id pubmed-10544308
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher PeerJ Inc.
record_format MEDLINE/PubMed
spelling pubmed-105443082023-10-03 Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice Qiu, Yuxiang Kang, Ningsu Wang, Xi Yao, Yao Cui, Jun Zhang, Xiaoyan Zheng, Lu PeerJ Biochemistry BACKGROUND: End-stage renal disease is primarily caused by diabetic kidney disease (DKD). The Farnesoid X receptor (FXR), a member of the nuclear receptor superfamily, has anti-inflammatory, lipid-lowering and hypoglycemic properties. It also inhibits renal fibrosis. Although its physiological role is not fully understood, it also plays a role in the control of diabetic nephropathy (DN). METHODS: In the present study, we examined male FXR & leptin receptor double knockout mice, in which weight, blood glucose, body fat, and other indicators were monitored. After 6 months of rearing, blood and urine samples were collected and biochemical parameters were measured. Fibrosis was assessed by Masson’s stain, while the assessment of the resuscitation case’s condition was performed using succinate dehydrogenase (SDHA) stain immunohistochemistry, which measures aerobic respiration. Expression of molecules such as connective tissue growth factor (CTGF), SMAD family members 3 (Smad3) and 7 (Smad7), and small heterodimer partner were detected by RT-PCR and Western blotting as part of the application. RESULTS: FXR knockout decreased body weight and body fat in db/db mice, but increased blood glucose, urine output, and renal fibrosis. Primary mesangial cells (P-MCs) from FXR(+/ +) mice stimulated with transforming growth factor β1 (TGFβ1) showed significantly higher levels of related fibrosis factors, TGFβ1 and Smad3 mRNA and protein, and significantly reduced levels of Smad7. These effects were reversed by the action of FXR agonist chenodeoxycholic acid (CDCA). P-MCs from FXR(−/ −) mice stimulated with TGFβ1 resulted in an increase in the expression and protein levels of collagen I and TGFβ1, and the addition of CDCA had no significant effect on TGFβ1 stimulation. However, compared with FXR(+/ +)db/db mice, the rate of oxygen consumption, the rate of carbon dioxide production, and the rate of energy conversion were increased in FXR(−/ −)db/db mice, whereas the SDHA succinate dehydrogenase, a marker enzyme for aerobic respiration, was significantly decreased. CONCLUSIONS: These results provide evidence that FXR plays a critical role in the regulation of mesangial cells in DN. The likely mechanism is that aberrant FXR expression activates TGFβ1, which induces extracellular matrix accumulation through the classical Smad signaling pathway, leading to mitochondrial dysfunction. PeerJ Inc. 2023-09-29 /pmc/articles/PMC10544308/ /pubmed/37790634 http://dx.doi.org/10.7717/peerj.16155 Text en ©2023 Qiu 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, reproduction and adaptation in any medium and for any purpose provided that it is properly attributed. For attribution, the original author(s), title, publication source (PeerJ) and either DOI or URL of the article must be cited.
spellingShingle Biochemistry
Qiu, Yuxiang
Kang, Ningsu
Wang, Xi
Yao, Yao
Cui, Jun
Zhang, Xiaoyan
Zheng, Lu
Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title_full Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title_fullStr Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title_full_unstemmed Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title_short Loss of Farnesoid X receptor (FXR) accelerates dysregulated glucose and renal injury in db/db mice
title_sort loss of farnesoid x receptor (fxr) accelerates dysregulated glucose and renal injury in db/db mice
topic Biochemistry
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10544308/
https://www.ncbi.nlm.nih.gov/pubmed/37790634
http://dx.doi.org/10.7717/peerj.16155
work_keys_str_mv AT qiuyuxiang lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT kangningsu lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT wangxi lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT yaoyao lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT cuijun lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT zhangxiaoyan lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice
AT zhenglu lossoffarnesoidxreceptorfxracceleratesdysregulatedglucoseandrenalinjuryindbdbmice