Cargando…

Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice

INTRODUCTION: Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS)...

Descripción completa

Detalles Bibliográficos
Autores principales: Favor, Olivia K., Chauhan, Preeti S., Pourmand, Elham, Edwards, Angel M., Wagner, James G., Lewandowski, Ryan P., Heine, Lauren K., Harkema, Jack R., Lee, Kin Sing Stephen, Pestka, James J.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9978350/
https://www.ncbi.nlm.nih.gov/pubmed/36875087
http://dx.doi.org/10.3389/fimmu.2023.1124910
_version_ 1784899503442100224
author Favor, Olivia K.
Chauhan, Preeti S.
Pourmand, Elham
Edwards, Angel M.
Wagner, James G.
Lewandowski, Ryan P.
Heine, Lauren K.
Harkema, Jack R.
Lee, Kin Sing Stephen
Pestka, James J.
author_facet Favor, Olivia K.
Chauhan, Preeti S.
Pourmand, Elham
Edwards, Angel M.
Wagner, James G.
Lewandowski, Ryan P.
Heine, Lauren K.
Harkema, Jack R.
Lee, Kin Sing Stephen
Pestka, James J.
author_sort Favor, Olivia K.
collection PubMed
description INTRODUCTION: Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS) which is devoid of O-antigen polysaccharide sidechain. Since these chemotypes differentially affect toll-like receptor 4 (TLR4)-mediated immune cell responses, these differences may influence GN induction. METHODS: We initially compared the effects of subchronic intraperitoneal (i.p.) injection for 5 wk with 1) Salmonella S-LPS, 2) Salmonella R-LPS, or 3) saline vehicle (VEH) (Study 1) in female NZBWF1 mice. Based on the efficacy of R-LPS in inducing GN, we next used it to compare the impact of two lipidome-modulating interventions, ω-3 polyunsaturated fatty acid (PUFA) supplementation and soluble epoxide hydrolase (sEH) inhibition, on GN (Study 2). Specifically, effects of consuming ω-3 docosahexaenoic acid (DHA) (10 g/kg diet) and/or the sEH inhibitor 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl) urea (TPPU) (22.5 mg/kg diet ≈ 3 mg/kg/day) on R-LPS triggering were compared. RESULTS: In Study 1, R-LPS induced robust elevations in blood urea nitrogen, proteinuria, and hematuria that were not evident in VEH- or S-LPS-treated mice. R-LPS-treated mice further exhibited kidney histopathology including robust hypertrophy, hyperplasia, thickened membranes, lymphocytic accumulation containing B and T cells, and glomerular IgG deposition consistent with GN that was not evident in VEH- or SLPS-treated groups. R-LPS but not S-LPS induced spleen enlargement with lymphoid hyperplasia and inflammatory cell recruitment in the liver. In Study 2, resultant blood fatty acid profiles and epoxy fatty acid concentrations reflected the anticipated DHA- and TPPU-mediated lipidome changes, respectively. The relative rank order of R-LPS-induced GN severity among groups fed experimental diets based on proteinuria, hematuria, histopathologic scoring, and glomerular IgG deposition was: VEH/CON< R-LPS/DHA ≈ R-LPS/TPPU<<< R-LPS/TPPU+DHA ≈ R-LPS/CON. In contrast, these interventions had modest-to- negligible effects on R-LPS-induced splenomegaly, plasma antibody responses, liver inflammation, and inflammation-associated kidney gene expression. DISCUSSION: We show for the first time that absence of O-antigenic polysaccharide in R-LPS is critical to accelerated GN in lupus-prone mice. Furthermore, intervention by lipidome modulation through DHA feeding or sEH inhibition suppressed R-LPS-induced GN; however, these ameliorative effects were greatly diminished upon combining the treatments.
format Online
Article
Text
id pubmed-9978350
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-99783502023-03-03 Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice Favor, Olivia K. Chauhan, Preeti S. Pourmand, Elham Edwards, Angel M. Wagner, James G. Lewandowski, Ryan P. Heine, Lauren K. Harkema, Jack R. Lee, Kin Sing Stephen Pestka, James J. Front Immunol Immunology INTRODUCTION: Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS) which is devoid of O-antigen polysaccharide sidechain. Since these chemotypes differentially affect toll-like receptor 4 (TLR4)-mediated immune cell responses, these differences may influence GN induction. METHODS: We initially compared the effects of subchronic intraperitoneal (i.p.) injection for 5 wk with 1) Salmonella S-LPS, 2) Salmonella R-LPS, or 3) saline vehicle (VEH) (Study 1) in female NZBWF1 mice. Based on the efficacy of R-LPS in inducing GN, we next used it to compare the impact of two lipidome-modulating interventions, ω-3 polyunsaturated fatty acid (PUFA) supplementation and soluble epoxide hydrolase (sEH) inhibition, on GN (Study 2). Specifically, effects of consuming ω-3 docosahexaenoic acid (DHA) (10 g/kg diet) and/or the sEH inhibitor 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl) urea (TPPU) (22.5 mg/kg diet ≈ 3 mg/kg/day) on R-LPS triggering were compared. RESULTS: In Study 1, R-LPS induced robust elevations in blood urea nitrogen, proteinuria, and hematuria that were not evident in VEH- or S-LPS-treated mice. R-LPS-treated mice further exhibited kidney histopathology including robust hypertrophy, hyperplasia, thickened membranes, lymphocytic accumulation containing B and T cells, and glomerular IgG deposition consistent with GN that was not evident in VEH- or SLPS-treated groups. R-LPS but not S-LPS induced spleen enlargement with lymphoid hyperplasia and inflammatory cell recruitment in the liver. In Study 2, resultant blood fatty acid profiles and epoxy fatty acid concentrations reflected the anticipated DHA- and TPPU-mediated lipidome changes, respectively. The relative rank order of R-LPS-induced GN severity among groups fed experimental diets based on proteinuria, hematuria, histopathologic scoring, and glomerular IgG deposition was: VEH/CON< R-LPS/DHA ≈ R-LPS/TPPU<<< R-LPS/TPPU+DHA ≈ R-LPS/CON. In contrast, these interventions had modest-to- negligible effects on R-LPS-induced splenomegaly, plasma antibody responses, liver inflammation, and inflammation-associated kidney gene expression. DISCUSSION: We show for the first time that absence of O-antigenic polysaccharide in R-LPS is critical to accelerated GN in lupus-prone mice. Furthermore, intervention by lipidome modulation through DHA feeding or sEH inhibition suppressed R-LPS-induced GN; however, these ameliorative effects were greatly diminished upon combining the treatments. Frontiers Media S.A. 2023-02-16 /pmc/articles/PMC9978350/ /pubmed/36875087 http://dx.doi.org/10.3389/fimmu.2023.1124910 Text en Copyright © 2023 Favor, Chauhan, Pourmand, Edwards, Wagner, Lewandowski, Heine, Harkema, Lee and Pestka 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 Immunology
Favor, Olivia K.
Chauhan, Preeti S.
Pourmand, Elham
Edwards, Angel M.
Wagner, James G.
Lewandowski, Ryan P.
Heine, Lauren K.
Harkema, Jack R.
Lee, Kin Sing Stephen
Pestka, James J.
Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title_full Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title_fullStr Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title_full_unstemmed Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title_short Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice
title_sort lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough lps-accelerated glomerulonephritis in lupus-prone mice
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9978350/
https://www.ncbi.nlm.nih.gov/pubmed/36875087
http://dx.doi.org/10.3389/fimmu.2023.1124910
work_keys_str_mv AT favoroliviak lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT chauhanpreetis lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT pourmandelham lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT edwardsangelm lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT wagnerjamesg lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT lewandowskiryanp lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT heinelaurenk lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT harkemajackr lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT leekinsingstephen lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice
AT pestkajamesj lipidomemodulationbydietaryomega3polyunsaturatedfattyacidsupplementationorselectivesolubleepoxidehydrolaseinhibitionsuppressesroughlpsacceleratedglomerulonephritisinlupuspronemice