Abstract

The hormone, relaxin, inhibits aberrant myofibroblast differentiation and collagen deposition by disrupting the TGF-β1/Smad2 axis, via its cognate receptor, Relaxin Family Peptide Receptor 1 (RXFP1), extracellular signal-regulated kinase (ERK)1/2 phosphorylation (pERK) and a neuronal nitric oxide (NO) synthase (nNOS)-NO-cyclic guanosine monophosphate (cGMP)-dependent pathway. However, the signalling pathways involved in its additional ability to increase matrix metalloproteinase (MMP) expression and activity remain unknown. This study investigated the extent to which the NO pathway was involved in human gene-2 (H2) relaxin's ability to positively regulate MMP-1 and its rodent orthologue, MMP-13, MMP-2 and MMP-9 (the main collagen-degrading MMPs) in TGF-β1-stimulated human dermal fibroblasts and primary renal myofibroblasts isolated from injured rats; by gelatin zymography (media) and Western blotting (cell layer). H2 relaxin (10–100 ng/ml) significantly increased MMP-1 (by ∼50%), MMP-2 (by ∼80%) and MMP-9 (by ∼80%) in TGF-β1-stimulated human dermal fibroblasts; and MMP-13 (by ∼90%), MMP-2 (by ∼130%) and MMP-9 (by ∼115%) in rat renal myofibroblasts (all p<0.01 vs untreated cells) over 72 hours. The relaxin-induced up-regulation of these MMPs, however, was significantly blocked by a non-selective NOS inhibitor (L-nitroarginine methyl ester (hydrochloride); L-NAME; 75–100 µM), and specific inhibitors to nNOS (N-propyl-L-arginine; NPLA; 0.2–2 µM), iNOS (1400W; 0.5–1 µM) and guanylyl cyclase (ODQ; 5 µM) (all p<0.05 vs H2 relaxin alone), but not eNOS (L-N-(1-iminoethyl)ornithine dihydrochloride; L-NIO; 0.5–5 µM). However, neither of these inhibitors affected basal MMP expression at the concentrations used. Furthermore, of the NOS isoforms expressed in renal myofibroblasts (nNOS and iNOS), H2 relaxin only stimulated nNOS expression, which in turn, was blocked by the ERK1/2 inhibitor (PD98059; 1 µM). These findings demonstrated that H2 relaxin signals through a RXFP1-pERK-nNOS-NO-cGMP-dependent pathway to mediate its anti-fibrotic actions, and additionally signals through iNOS to up-regulate MMPs; the latter being suppressed by TGF-β1 in myofibroblasts, but released upon H2 relaxin-induced inhibition of the TGF-β1/Smad2 axis.

Highlights

  • Fibrosis is a universal response to chronic injury and inflammation in several organs and its failure to resolve leads to significant dysfunction and onset of organ failure [1,2]

  • human gene-2 (H2) relaxin did not have any effects on iNOS expression, upon administration to these cells (Figure 4B); implying that relaxin was primarily signalling through a RXFP1-pERK-nitric oxide (NO) synthase (nNOS)-nitric oxide (NO)-cGMPdependent pathway to positively regulate matrix metalloproteinase (MMP); but that iNOS was somehow involved at a down-stream level

  • We demonstrate for the first time in transforming growth factor (TGF)-b1-stimulated human dermal fibroblasts and rat renal myofibroblasts that express RXFP1, that relaxin primarily signals through this RXFP1-pERK-nNOS-NO-cyclic guanosine monophosphate (cGMP)-dependent pathway to up-regulate MMP-1, MMP-2 and MMP-9; which are known to degrade collagens and other matrix proteins [26,44,45,46,47]

Read more

Summary

Introduction

Fibrosis is a universal response to chronic injury and inflammation in several organs and its failure to resolve leads to significant dysfunction and onset of organ failure [1,2]. Excessive collagen deposition (the main constituent of fibrotic tissue) leads to adverse outcomes, with damage depending on the quantity of matrix produced (fibrogenesis), and the degree of its cross-linking and its reorganisation, or density. Fibrosis is dependent to a large extent on the recruitment of myofibroblasts, cells with the phenotypic features of both fibroblasts and vascular smooth muscle [3]. Recognised by their de novo expression of a smooth muscle actin (aSMA), myofibroblasts are prodigious producers of the ECM and are influenced by several mediators, including cytokines, chemokines and growth factors [3,4]. Expression of TGF-b1 can be induced by mechanical overload, myocardial ischemia, cardiomyopathy or angiotensin II (Ang II) [5,6]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call