THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE

INTRODUCTION: The anti-fibrotic hormone, relaxin, has been inferred to disrupt TGF-beta1/Smad2 phosphorylation (pSmad2) signal transduction and promote collagen-degrading gelatinase activity via a nitric oxide (NO)-dependent pathway. Here, we determined the extent to which NO, soluble guanylate cycl...

Full description

Bibliographic Details
Main Authors: Chao eWang, Barbara Kathryn Kemp-Harper, Martina eKocan, Sheng Yu eAng, Tim David Hewitson, Chrishan S Samuel
Format: Article
Language:English
Published: Frontiers Media S.A. 2016-03-01
Series:Frontiers in Pharmacology
Subjects:
Online Access:http://journal.frontiersin.org/Journal/10.3389/fphar.2016.00091/full
id doaj-067a8e7a4f664c2bae20e57d81baaff9
record_format Article
spelling doaj-067a8e7a4f664c2bae20e57d81baaff92020-11-24T22:25:12ZengFrontiers Media S.A.Frontiers in Pharmacology1663-98122016-03-01710.3389/fphar.2016.00091190302THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATEChao eWang0Barbara Kathryn Kemp-Harper1Martina eKocan2Sheng Yu eAng3Tim David Hewitson4Chrishan S Samuel5Monash UniversityMonash UniversityDrug Discovery Biology, Monash Institute of Pharmacurtical Sciences, Monash UniversityDrug Discovery Biology, Monash Institute of Pharmacurtical Sciences, Monash UniversityRoyal Melbourne HospitalMonash UniversityINTRODUCTION: The anti-fibrotic hormone, relaxin, has been inferred to disrupt TGF-beta1/Smad2 phosphorylation (pSmad2) signal transduction and promote collagen-degrading gelatinase activity via a nitric oxide (NO)-dependent pathway. Here, we determined the extent to which NO, soluble guanylate cyclase (sGC) and cyclic guanosine monophosphate (cGMP) were directly involved in the anti-fibrotic actions of relaxin using a selective NO scavenger and sGC inhibitor, and comparing and combining relaxin’s effects with that of an NO donor. METHODS AND RESULTS: Primary renal cortical myofibroblasts isolated from injured rat kidneys were treated with human recombinant relaxin (RLX; 16.8nM), the NO donor, diethylamine NONOate (DEA/NO; 0.5-5uM) or the combined effects of RLX (16.8nM) and DEA/NO (5uM) over 72 hours. The effects of RLX (16.8nM) and DEA/NO (5uM) were also evaluated in the presence of the NO scavenger, hydroxocobalamin (HXC; 100uM) or sGC inhibitor, ODQ (5uM) over 72 hours. Furthermore, the effects of RLX (30nM), DEA/NO (5uM) and RLX (30nM)+DEA/NO (5uM) on cGMP levels were directly measured, in the presence or absence of ODQ (5uM). Changes in matrix metalloproteinase (MMP)-2, MMP-9 (cell media), pSmad2 and α-smooth muscle actin (α-SMA; a measure myofibroblast differentiation) (cell layer) were assessed by gelatin zymography and Western blotting, respectively. At the highest concentration tested, both RLX and DEA/NO promoted MMP-2 and MMP-9 levels by 25-33%, while inhibiting pSmad2 and α-SMA expression by up to 50% (all p<0.05 vs untreated and vehicle-treated cells). However, 5uM of DEA/NO was required to produce the effects seen with 16.8nM of RLX over 72 hours. The anti-fibrotic effects of RLX or DEA/NO alone were completely abrogated by HXC and ODQ (both p<0.01 vs RLX alone or DEA/NO alone), but were significantly enhanced when added in combination (all p<0.05 vs RLX alone). Additionally, the direct cGMP-promoting effects of RLX, DEA/NO and RLX+DEA/NO (which all increased cGMP levels by 12-16-fold over basal levels; all p<0.01 vs vehicle-treated cells) were significantly inhibited by pre-treatment of ODQ (all p<0.05 vs the respective treatments alone). CONCLUSIONS: These findings confirmed that RLX mediates its TGF-beta1-inhibitory and gelatinase-promoting effects via a NO-sGC-cGMP-dependent pathway, which was additively augmented by co-administration of DEA/NO.http://journal.frontiersin.org/Journal/10.3389/fphar.2016.00091/fullFibrosisMyofibroblastsNitric OxideRelaxinCell signalingcGMP
collection DOAJ
language English
format Article
sources DOAJ
author Chao eWang
Barbara Kathryn Kemp-Harper
Martina eKocan
Sheng Yu eAng
Tim David Hewitson
Chrishan S Samuel
spellingShingle Chao eWang
Barbara Kathryn Kemp-Harper
Martina eKocan
Sheng Yu eAng
Tim David Hewitson
Chrishan S Samuel
THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
Frontiers in Pharmacology
Fibrosis
Myofibroblasts
Nitric Oxide
Relaxin
Cell signaling
cGMP
author_facet Chao eWang
Barbara Kathryn Kemp-Harper
Martina eKocan
Sheng Yu eAng
Tim David Hewitson
Chrishan S Samuel
author_sort Chao eWang
title THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
title_short THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
title_full THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
title_fullStr THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
title_full_unstemmed THE ANTI-FIBROTIC ACTIONS OF RELAXIN ARE MEDIATED THROUGH A NO-sGC-cGMP-DEPENDENT PATHWAY IN RENAL MYOFIBROBLASTS IN VITRO AND ENHANCED BY THE NO DONOR, DIETHYLAMINE NONOATE
title_sort anti-fibrotic actions of relaxin are mediated through a no-sgc-cgmp-dependent pathway in renal myofibroblasts in vitro and enhanced by the no donor, diethylamine nonoate
publisher Frontiers Media S.A.
series Frontiers in Pharmacology
issn 1663-9812
publishDate 2016-03-01
description INTRODUCTION: The anti-fibrotic hormone, relaxin, has been inferred to disrupt TGF-beta1/Smad2 phosphorylation (pSmad2) signal transduction and promote collagen-degrading gelatinase activity via a nitric oxide (NO)-dependent pathway. Here, we determined the extent to which NO, soluble guanylate cyclase (sGC) and cyclic guanosine monophosphate (cGMP) were directly involved in the anti-fibrotic actions of relaxin using a selective NO scavenger and sGC inhibitor, and comparing and combining relaxin’s effects with that of an NO donor. METHODS AND RESULTS: Primary renal cortical myofibroblasts isolated from injured rat kidneys were treated with human recombinant relaxin (RLX; 16.8nM), the NO donor, diethylamine NONOate (DEA/NO; 0.5-5uM) or the combined effects of RLX (16.8nM) and DEA/NO (5uM) over 72 hours. The effects of RLX (16.8nM) and DEA/NO (5uM) were also evaluated in the presence of the NO scavenger, hydroxocobalamin (HXC; 100uM) or sGC inhibitor, ODQ (5uM) over 72 hours. Furthermore, the effects of RLX (30nM), DEA/NO (5uM) and RLX (30nM)+DEA/NO (5uM) on cGMP levels were directly measured, in the presence or absence of ODQ (5uM). Changes in matrix metalloproteinase (MMP)-2, MMP-9 (cell media), pSmad2 and α-smooth muscle actin (α-SMA; a measure myofibroblast differentiation) (cell layer) were assessed by gelatin zymography and Western blotting, respectively. At the highest concentration tested, both RLX and DEA/NO promoted MMP-2 and MMP-9 levels by 25-33%, while inhibiting pSmad2 and α-SMA expression by up to 50% (all p<0.05 vs untreated and vehicle-treated cells). However, 5uM of DEA/NO was required to produce the effects seen with 16.8nM of RLX over 72 hours. The anti-fibrotic effects of RLX or DEA/NO alone were completely abrogated by HXC and ODQ (both p<0.01 vs RLX alone or DEA/NO alone), but were significantly enhanced when added in combination (all p<0.05 vs RLX alone). Additionally, the direct cGMP-promoting effects of RLX, DEA/NO and RLX+DEA/NO (which all increased cGMP levels by 12-16-fold over basal levels; all p<0.01 vs vehicle-treated cells) were significantly inhibited by pre-treatment of ODQ (all p<0.05 vs the respective treatments alone). CONCLUSIONS: These findings confirmed that RLX mediates its TGF-beta1-inhibitory and gelatinase-promoting effects via a NO-sGC-cGMP-dependent pathway, which was additively augmented by co-administration of DEA/NO.
topic Fibrosis
Myofibroblasts
Nitric Oxide
Relaxin
Cell signaling
cGMP
url http://journal.frontiersin.org/Journal/10.3389/fphar.2016.00091/full
work_keys_str_mv AT chaoewang theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT barbarakathrynkempharper theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT martinaekocan theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT shengyueang theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT timdavidhewitson theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT chrishanssamuel theantifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT chaoewang antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT barbarakathrynkempharper antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT martinaekocan antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT shengyueang antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT timdavidhewitson antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
AT chrishanssamuel antifibroticactionsofrelaxinaremediatedthroughanosgccgmpdependentpathwayinrenalmyofibroblastsinvitroandenhancedbythenodonordiethylaminenonoate
_version_ 1725758881600110592