Academia.eduAcademia.edu

Anti-fibrotic actions of relaxin

2016, British journal of pharmacology

Fibrosis refers to the hardening or scarring of tissues that usually results from aberrant wound healing in response to organ injury, and its manifestations in various organs have collectively been estimated to contribute to around 45-50% of deaths in the Western world. Despite this, there is currently no effective cure for the tissue structural and functional damage induced by fibrosis-related disorders. Relaxin meets several criteria of an effective anti-fibrotic based on its specific ability to inhibit pro-fibrotic cytokine and/or growth factor-mediated, but not normal/unstimulated, fibroblast proliferation, differentiation and matrix production. Furthermore, relaxin augments matrix degradation through its ability to up-regulate the release and activation of various matrix-degrading matrix metalloproteinases and/or being able to down-regulate tissue inhibitor of metalloproteinase activity. Relaxin can also indirectly suppress fibrosis through its other well-known (anti-inflammato...

BJP British Journal of Pharmacology British Journal of Pharmacology (2017) 174 962–976 962 Themed Section: Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors REVIEW ARTICLE Anti-fibrotic actions of relaxin Correspondence Associate Professor Chrishan S. Samuel, PhD, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Vic. 3800, Australia; and Professor Robert G. Bennett, PhD, Research Service 151 VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA. E-mail: chrishan.samuel@monash.edu; rgbennet@unmc.edu Received 7 March 2016; Revised 19 May 2016; Accepted 23 May 2016 C S Samuel1, S G Royce1, T D Hewitson3, K M Denton2, T E Cooney4 and R G Bennett5,6 1 Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Melbourne, Vic., Australia, 2 Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Vic., Australia, Department of Nephrology, Royal Melbourne Hospital, Melbourne, Vic., Australia, 4University of Pittsburgh Medical Centre (UPMC) Hamot, Erie, PA, 3 USA, 5Research Service 151, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA, and 6Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA Fibrosis refers to the hardening or scarring of tissues that usually results from aberrant wound healing in response to organ injury, and its manifestations in various organs have collectively been estimated to contribute to around 45–50% of deaths in the Western world. Despite this, there is currently no effective cure for the tissue structural and functional damage induced by fibrosisrelated disorders. Relaxin meets several criteria of an effective anti-fibrotic based on its specific ability to inhibit pro-fibrotic cytokine and/or growth factor-mediated, but not normal/unstimulated, fibroblast proliferation, differentiation and matrix production. Furthermore, relaxin augments matrix degradation through its ability to up-regulate the release and activation of various matrix-degrading matrix metalloproteinases and/or being able to down-regulate tissue inhibitor of metalloproteinase activity. Relaxin can also indirectly suppress fibrosis through its other well-known (anti-inflammatory, antioxidant, antihypertrophic, anti-apoptotic, angiogenic, wound healing and vasodilator) properties. This review will outline the organ-specific and general anti-fibrotic significance of exogenously administered relaxin and its mechanisms of action that have been documented in various non-reproductive organs such as the cardiovascular system, kidney, lung, liver, skin and tendons. In addition, it will outline the influence of sex on relaxin’s anti-fibrotic actions, highlighting its potential as an emerging anti-fibrotic therapeutic. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc Abbreviations α-SMA, α-smooth muscle actin; AT2 receptor, angiotensin type 2 receptor; CCl4, carbon tetrachloride; ECM, extracellular matrix; HSC, hepatic stellate cell; INSL, insulin-like; KO, knockout; MSC, mesenchymal stem cell; PMA, phorbol 12-myristate 13-acetate; RLN, relaxin gene; rhRLX, synthetically or recombinantly produced drug form of relaxin; RXFP1, relaxin family peptide receptor 1; TIMP, tissue inhibitor of metalloproteinase DOI:10.1111/bph.13529 © 2016 The British Pharmacological Society Anti-fibrotic actions of relaxin BJP Tables of Links TARGETS Other protein targets LIGANDS a Enzymes VEGF GPCRs b d Angiotensin II Relaxin Akt (PKB) cAMP TGF-β1 MMP-1 Enalapril TIMP1 TIMP2 β2-adrenoceptor MMP-2 IFN-γ AT2 receptor MMP-9 Methylprednisolone RXFP1 receptor MMP-13 RXFP2 receptor Nuclear hormone receptors nNOS c PKA PPARγ These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Southan et al., 2016) and are permanently archived in the Concise Guide a,b,c,d Alexander et al., 2015a,b,c,d). to PHARMACOLOGY 2015/16 ( Introduction The tissue response to stress depends on a number of intrinsic and extrinsic factors including the length and extent of injury. When injury or disease is mild or transient, then the tissue response leads to remodelling or regeneration of the organ parenchyma. However, if the injury is either severe or prolonged, the process is characterized by ongoing inflammation and extracellular matrix (ECM) production. Fibrosis is therefore a failure of the wound healing process, where ECM synthesis is ongoing (Wynn, 2008; Hewitson, 2009). This maladaptive response is particularly dependent on paracrine and autocrine production of TGF-β1 and the consequent recruitment of activated fibroblasts (myofibroblasts). Over-abundant ECM production and failure to resolve lead to significant organ damage and dysfunction, which depends not only on the quantity of matrix produced (fibrogenesis) but also the degree of its cross-linking and its reorganization, or density. Despite the almost universal significance of fibrosis (Wynn, 2008; Hewitson, 2009), there is a lack of effective anti-fibrotic therapeutic strategies. A growing body of evidence suggests that relaxin may fulfil this need. This review will focus on its anti-fibrotic effects and mechanism of action in tissues of the circulatory, renal, hepatic and integumentary systems. Moreover, the influence of sex on fibrosis progression will be discussed. Relaxin Relaxin is a member of a family of peptide hormones that is structurally similar to insulin, but which diverged from insulin early in vertebrate evolution to form a distinct peptide family based on their two-chain structures, receptor binding motif and ability to bind and activate GPCRs (refer to pp. 5846–7 in Alexander et al., 2015b; Bathgate et al., 2006), as opposed to the tyrosine kinase receptors that are activated by insulin and the insulin-like growth factors. The relaxin peptide family is encoded by seven genes in humans, which include the relaxin genes RLN1, RLN2 and RLN3, as well as the insulin-like peptide genes INSL3, INSL4, INSL5 and INSL6 (Bathgate et al., 2006). In most other species though, only five of these genes exist, including RLN1 and RLN3, the species equivalents of human RLN2 and RLN3 respectively. The product of the human RLN2 gene (H2 relaxin) and its species equivalent gene (relaxin) represent the major stored and circulating forms of relaxin in their respective species and will be the forms of relaxin primarily discussed in this review. Recombinant human relaxin [synthetically or recombinantly produced drug form of relaxin (rhRLX), now also referred to as serelaxin] is the drug-based form of H2 relaxin, which will also be discussed throughout the review. Anti-fibrotic effects of relaxin in the cardiovascular system Fibrosis is a hallmark of almost all forms of cardiovascular disease and a key contributor to atrial, ventricular and vascular stiffness, impaired cardiac contractility and heart failure (Mandavia et al., 2013; Frieler and Mortensen, 2015; Ling et al., 2016). A common fibrotic process develops from a number of cardiac pathologies including ischaemic injury and myocardial infarction; hypertrophic, dilated and restrictive cardiomyopathies; valvular heart diseases; hypertension; diabetes; metabolic disorders; and cardiac arrhythmia, amongst others. Continuous exogenous administration of rhRLX, human gene-3 relaxin (H3 RLX) or mouse relaxin (mRLX) to profibrotic factor-stimulated cardiac fibroblasts in vitro (at 100 ng·mL 1 over 1–5 days) or various experimental models of cardiovascular disease in vivo (at 0.1–0.5 mg·kg 1·day 1 over 2–4 weeks, where 0.5 mg·kg 1·day 1 produces circulating levels of ~20–40 ng·mL 1) has effectively reduced the cardiac fibrosis associated with each model investigated (Table 1). The anti-fibrotic effects of rhRLX have consistently been demonstrated in various preclinical models of British Journal of Pharmacology (2017) 174 962–976 963 BJP C S Samuel et al. Table 1 Various models of cardiovascular disease that have been used to demonstrate the anti-fibrotic effects of relaxin Model studied Type of relaxin used Length of treatment (days) Ang II-stimulated atrial fibroblasts (rat) (Samuel et al., 2004a) rhRLX 3 Ang II-stimulated cardiac fibroblasts (rat) (Gu et al., 2012) rhRLX 1 TGF-β1-stimulated atrial fibroblasts (rat) (Samuel et al., 2004a) rhRLX 3 TGF-β1-stimulated ventricular fibroblasts (rat) (Samuel et al., 2004a) rhRLX 3 TGF-β1-stimulated cardiac fibroblasts (mouse) (Sassoli et al., 2013; Squecco et al., 2015) rhRLX 1–5 PMA-stimulated cardiac fibroblasts (rat) (Wang et al., 2014) rhRLX 3 High glucose-stimulated cardiac fibroblasts (rat) (Wang et al., 2009; Su et al., 2014) rhRLX 2 Age-related fibrosis (mouse) (Samuel et al., 2004a) (Samuel et al., 2007)a rhRLX/mRLXa 14/120a β2-adrenoreceptor-induced cardiomyopathy (mouse) # (Samuel et al., 2004a; Bathgate et al., 2008) (Hossain et al., 2011a)c (Chan et al., 2012; Samuel et al., 2014) Ad-mRLXb/H3 RLXc/rhRLX 14 Myocardial infarction (pig)(Formigli et al., 2007)d, (rat) (Bonacchi et al., 2009), (mouse) (Samuel et al., 2011) hRLXd/rhRLX 7–30 Hypertension (rat) (Lekgabe et al., 2005; Gu et al., 2012) rhRLX 14 Type 1 diabetes (rat) (Samuel et al., 2008) rhRLX 14 Atrial fibrillation (rat) (Parikh et al., 2013; Henry et al., 2016) rhRLX 14 a Synthetically produced mRLX. b adenovirus-mediated mRLX (Ad-mRLX). c H3 RLX was used. d mouse skeletal myoblasts were engineered to produce relaxin based on the hRLX sequence. cardiovascular disease and heart failure in vivo, regardless of aetiology, including models of ageing (Samuel et al., 2007; Samuel et al., 2004a), fibrotic cardiomyopathy (Chan et al., 2012; Samuel et al., 2014), myocardial infarction (Formigli et al., 2007; Bonacchi et al., 2009; Samuel et al., 2011), hypertension (Lekgabe et al., 2005; Gu et al., 2012), type 1 diabetes (Samuel et al., 2008) and atrial fibrillation (Parikh et al., 2013; Henry et al., 2016) (Table 1). Furthermore, rhRLX (0.5 mg·kg 1·day 1) was found to have improved anti-fibrotic efficacy over the clinically used ACE inhibitor, enalapril (48 mg·kg 1·day 1), in an experimental model of fibrotic cardiomyopathy, but also augmented the anti-fibrotic efficacy of enalapril when both treatments were administered 10 days after the onset of injury (Samuel et al., 2014). Likewise, both H3 RLX (Hossain et al., 2011a) and mRLX (Samuel et al., 2007; Bathgate et al., 2008) have also demonstrated similar anti-fibrotic efficacy to that of hRLX in murine models of cardiomyopathy induced by transgenic overexpression of β2-adrenoceptors. All forms of relaxin studied to date mediate their anti-fibrotic actions by inhibiting the effects of various pro-fibrotic factors (discussed below) rather than by directly regulating collagen or other ECM proteins per se, and rhRLX demonstrated greater anti-fibrotic efficacy than enalapril due to its greater ability to inhibit both TGF-β1 expression and its signal transduction at the level of intracellular Smad2 phosphorylation (Samuel et al., 2014). However, rhRLX did not affect chronic pressure overloadinduced cardiac hypertrophy or fibrosis that was associated with biochemical wall stress rather than elevated TGF-β1 levels (Xu et al., 2008). 964 British Journal of Pharmacology (2017) 174 962–976 Relevant mechanisms of the anti-fibrotic actions of relaxin Several studies conducted at the in vitro level have offered more in-depth insights into the mechanisms and signal transduction pathways associated with relaxin’s anti-fibrotic actions in the diseased myocardium, primarily involving rhRLX. rhRLX (and H3 RLX; Hossain et al., 2011a) acting via the relaxin family peptide receptor 1 (RXFP1 receptor) specifically inhibits cardiac fibroblast proliferation and differentiation into activated myofibroblasts and, hence, myofibroblast-mediated aberrant collagen deposition (the basis of fibrosis) when cells are stimulated with either angiotensin II (Samuel et al., 2004a; Gu et al., 2012), TGF-β1 (Samuel et al., 2004a; Sassoli et al., 2013; Squecco et al., 2015), phorbol 12-myristate 13-acetate (PMA; Wang et al., 2014) or high glucose (Wang et al., 2009; Su et al., 2014). The ability of rhRLX to inhibit TGF-β1-mediated cardiac fibrosis progression was shown to involve activation of the Notch-1 and NO pathways, and down-regulation of Smad2 (Samuel et al., 2014) and/or Smad3 (Sassoli et al., 2013) phosphorylation. This collectively results in the rhRLX-induced inhibition of the pro-fibrotic actions of TGF-β1 on myofibroblast differentiation and aberrant ECM/collagen deposition. Furthermore, by suppressing the TGF-β1/Smad2 and/or TGF-β1/Smad3 axes, which themselves promote fibrosis by inhibiting the actions of matrix-degrading MMPs and promoting tissue inhibitor of metalloproteinase (TIMP) activity (which are natural inhibitors of MMPs) (Spinale and Villarreal, 2014), rhRLX releases and promotes various MMPs (including MMP-1 and its rodent orthologue MMP-13, MMP-2 and MMP-9) (Lekgabe et al., 2005; Formigli Anti-fibrotic actions of relaxin et al., 2007; Parikh et al., 2013; Samuel et al., 2004a, 2008, 2011, 2014; Sarwar et al., 2015a,b) and/or decreases both cardiac TIMP-1 (Samuel et al., 2008) and TIMP-2 (Sassoli et al., 2013) activity, which would probably result in the degradation of existing collagen fibres. Hence, relaxin mediates its anti-fibrotic effects in the heart by suppressing aberrant profibrotic cytokine-induced ECM/collagen deposition, while also being able to promote MMP-induced ECM/collagen breakdown (Figure 1). rhRLX may also indirectly inhibit cardiac fibrosis through its anti-inflammatory (Nistri et al., 2003; Perna et al., 2005; 2008), antioxidant (Perna et al., 2005), anti-hypertrophic (Dschietzig et al., 2005; Moore et al., 2007; Parikh et al., 2013) and antiapoptotic (Moore et al., 2007; Bonacchi et al., 2009; Samuel et al., 2011; Zhang et al., 2015) actions, while being able to promote tissue repair via its angiogenic (Formigli et al., 2007; Samuel et al., 2011; Segal et al., 2012), vasodilatory (Conrad and Shroff, 2011; McGuane et al., 2011a,b) and wound healing BJP (Mu et al., 2010) properties. However, the effects of rhRLX have largely been shown to be independent of blood pressure regulation (Du et al., 2010; Samuel et al., 2008, 2014). These combined effects of rhRLX, along with its direct anti-fibrotic actions, have resulted in its ability to improve cardiac output and ventricular performance (Perna et al., 2005; Teichman et al., 2008), while being able to reduce cardiac contractility and ventricular stiffness (Samuel et al., 2008; Du et al., 2010) as well as atrial (Parikh et al., 2013) and ventricular (Nistri et al., 2008) arrhythmias. Anti-fibrotic actions of relaxin in the kidney The close association between cardiovascular pathology and renal dysfunction is well documented and significant. Patients with conventional risk factors for cardiovascular Figure 1 Summary of the mechanisms of relaxin’s anti-fibrotic actions that are mediated through RXFP1 receptors and RXFP1–AT2 receptor heterodimers. Relaxin specifically ameliorates the effects of pro-fibrotic stimuli such as TGF-β1 and Ang II, the former by inhibiting Smad2 (pSmad2) and/or Smad3 (pSmad3) phosphorylation, which is dependent at least in part on the pERK1/2, NO and Notch-1 pathways. This causes decreased expression and deposition of interstitial (types I, III and V) and basement membrane (type IV) collagens and reduced activity of TIMP-1 and TIMP-2, accompanied by increased expression and activity of various MMPs, including MMP-1/13, MMP-2 and/or MMP-9. The end result is a decrease in the rate of collagen deposition and increased collagen degradation, allowing clearance of the fibrotic scar. British Journal of Pharmacology (2017) 174 962–976 965 BJP C S Samuel et al. disease also suffer renal dysfunction. The pathology of the heart and kidney is therefore inexorably linked (Hewitson et al., 2015). Early studies showed that continuous 2–4 week infusion of rhRLX with osmotic mini-pumps ameliorated progressive fibrosis in several experimental rodent models of fibrosis including bromoethylamine-induced renal papillary necrosis (Garber et al., 2001), experimental anti-glomerular basement membrane nephritis (McDonald et al., 2003) and reduction of renal mass by either surgical ablation or infarction (Garber et al., 2003). The last two models are particularly insightful because they showed a decrease in hypertension in the infarction model, but no change in blood pressure in the normotensive ablation model, suggesting that structural–functional effects can be independent of changes in blood pressure (Garber et al., 2003). Also key is the observation that systemic administration of rhRLX concurrently limits both cardiac and renal fibrosis in the spontaneously hypertensive rat, indicating that rhRLX may simultaneously ameliorate similar pathologies in multiple organs (Lekgabe et al., 2005). Treatment of acute renal injury with rhRLX also limited loss of structure and function after ischaemia–reperfusion (Yoshida et al., 2013) and cisplatin exposure (Yoshida et al., 2014). However, rhRLX may not be universally renoprotective, as it did not prevent diabetic renal complications in mouse models of type I diabetes (Wong et al., 2013; Dschietzig et al., 2015), when TGF-β1 was not elevated in either model. Importantly, in a phase II scleroderma trial (Khanna et al., 2009), cessation of rhRLX after 24 weeks of administration resulted in a significant decline in renal function, as estimated by creatinine clearance (Khanna et al., 2009). It is unclear if this was related to scleroderma-specific factors. Establishing that pretreatment with rhRLX can slow progression is an important proof of principle, but the reality is that most renal patients present clinically with established fibrosis. It is those studies in established renal disease that will provide the greatest clinical insight. Relevant pleiotropic actions of relaxin The pleiotropic properties of rhRLX can also indirectly limit renal fibrosis and promote wound healing by stimulating NO production (Mookerjee et al., 2009; Sasser et al., 2011; Chow et al., 2012; Chow et al., 2014), improving the local environment through maintaining glomerular filtration rate (Conrad, 2004), preventing parenchymal cell loss through apoptosis (Yoshida et al., 2013) and enhancing cell survival through maintaining vascular supply via angiogenesis (Unemori et al., 2000; Hewitson et al., 2010). Its recognized direct cellular actions in the kidney include a reduction in inflammation (Yoshida et al., 2014), oxidative stress (Sasser et al., 2011) and ECM synthesis (fibrogenesis) and contraction (Masterson et al., 2004). There is also circumstantial evidence to suggest that a rhRLX-mediated increase in activity of MMP-2, MMP-9 or MMP-1 (and its analogue MMP-13 in the rodent) may directly limit renal scarring by increasing collagen degradation (Figure 1). Nevertheless, the spatial and temporal context of these collagenases are important as degradation of collagen IV can limit fibrosis by removing excess collagen and, in other circumstances, exacerbate injury through destruction of basement membranes. An 966 British Journal of Pharmacology (2017) 174 962–976 MMP-induced vasodilatation occurs through ET1–32 production (Jeyabalan et al., 2003), which again may improve wound repair and the environment in which the kidney functions. rhRLX may also indirectly reduce fibrosis by augmenting repair with mesenchymal stem cells (MSCs). A combination of rhRLX and bone marrow-derived MSCs attenuated kidney fibrosis after 7 days of unilateral ureteric obstruction (Huuskes et al., 2015). This was accompanied by reduced tissue damage and enhanced MMP-2 activity compared with either treatment alone, while rhRLX also directly increased MSC proliferation and migration (Huuskes et al., 2015). Signal transduction pathways of the anti-fibrotic actions of rhRLX Extensive cell culture studies using rat renal myofibroblasts have provided a mechanistic insight into rhRLX’s anti-fibrotic actions. rhRLX inhibits myofibroblast differentiation and collagen synthesis and promotes MMP expression/activity by myofibroblasts through binding to its cognate receptor, RXFP1 (Mookerjee et al., 2009). Recent studies also suggest that heterodimer formation between the RXFP1 receptor and other GPCRs, in particular angiotensin receptors, may be necessary (Chow et al., 2014). Regardless, subsequent signal transduction through a neuronal NOS–NO–cGMP pathway appears to be central to the anti-fibrotic actions of rhRLX in renal myofibroblasts. More specifically, rhRLX inhibits phosphorylation of Smad2 in both human (Heeg et al., 2005) and rat renal (myo)fibroblasts (Mookerjee et al., 2009). These effects are only partially dosedependent, with robust effects seen at physiological concentrations (Mookerjee et al., 2009). Importantly, the anti-fibrotic actions of rhRLX may well be contingent on TGF-β1, which may explain the lack of efficacy in models without aberrant TGF-β1 expression (Wong et al., 2013; Dschietzig et al., 2015). The influence of sex on the anti-fibrotic actions of relaxin There are sex differences in the incidence, prevalence and progression of cardiovascular and renal disease, with males being at greater risk than females prior to menopause (Silbiger and Neugarten, 2008; Barrett-Connor, 2013). For example, it has been demonstrated that cardiac fibrosis associated with left ventricular hypertrophy due to aortic stenosis is greater in men compared with women (Petrov et al., 2014). These data suggest that there are sex differences in the adaptation to pressure overload. Indeed, this greater cardiac fibrosis was associated with higher cardiac tissue levels of TGF-β1, Smad2 phosphorylation, and higher fibrosis related gene expression of collagens I and III, MMP-2 and MMP-9 in men than women (Petrov et al., 2010; Petrov et al., 2014). Similarly, in chronic kidney disease, tissue damage is greater in males than females, and this has been associated with enhanced TGF-β1 activation and reduced NO bioavailability (Silbiger and Neugarten, 2008). It is notable that these pathways are responsive to the actions of relaxin. Therefore, relaxin may act as a protective buffer against cardiovascular and renal disease in pre-menopausal women. Anti-fibrotic actions of relaxin The literature is unclear as to whether serum relaxin levels are greater in women than in men. In the main, data suggest that relaxin levels are greater in females during the luteal phase of the oestrous cycle and pregnancy (Bani, 2008). A recent study (Wolf et al., 2013) detected no difference in serum relaxin levels between men and women. However, given that the subjects were athletes, it is possible that ovulation and relaxin secretion was suppressed in these females. Thus, higher circulating levels of relaxin may confer cardio-renal protection during the reproductive years in women. Surprisingly few studies have examined whether relaxin has sexually dimorphic effects in ameliorating cardio-renal disease. Studies examining the response to rhRLX infusion observed no difference in the blood pressure, cardiac output, total peripheral resistance or renal function (Danielson et al., 2000; Conrad et al., 2004) between male and female rats. In other studies, the age-related cardiac and renal fibrosis observed in relaxin knockout (RLN1-KO) mice was exacerbated in male but not female mice (Du et al., 2003; Samuel et al., 2004b), which was later found to be associated with testosterone and elevated androgen receptor levels in RLN1-KO mice being causative of the problem (Hewitson et al., 2012). These data suggest that the effects of relaxin may be of greater importance in males than in females. However, an alternative explanation put forward was that females have additional protective mechanisms that compensate and mask the loss of relaxin (Du et al., 2003; Metra et al., 2013). Interestingly though, while neither ovariectomy or oestrogen replacement therapy affected the cardiac and renal fibrosis measured in female RLN1-KO mice (Lekgabe et al., 2006), ovariectomized RLN1-KO mice had exacerbated lung myofibroblast burden and subepithelial collagen levels (fibrosis), which were both diminished by oestrogen replacement therapy, suggesting that the potential compensation offered by oestrogen at least may be organ specific. In the Relaxin in Acute Heart Failure trial, subgroup analysis of the response to acute administration of rhRLX did not detect a sex difference in outcomes (Metra et al., 2013). However, systemic adenoviral delivery of mRLX attenuated left ventricular fibrosis in male and female β2-adrenoceptor transgenic mice (Bathgate et al., 2008), suggesting that rhRLX may be equally effective for the treatment of cardiovascular and renal disease in both sexes. However, more detailed studies examining the response to rhRLX administration in models of disease in both males and females are required to validate this. It would also be important to conduct studies in both young and aged animals, as cardiovascular and renal disease occur predominantly in post-menopausal women, when loss of ovarian hormones may reduce receptor and/or second messenger signalling pathways. There are also intriguing data to suggest that relaxin may have an enhanced action in females. As detailed above, rhRLX’s NO-promoting and TGF-β1 inhibitory actions were found to involve an interaction with the angiotensin type 2 receptor (AT2 receptor), through heterodimers formed between the RXFP1 and the AT2 receptors (Chow et al., 2012; 2014) (Figure 1). Moreover, we have demonstrated that the renal and vascular effects of AT2 receptor stimulation are enhanced in females in association with increased AT2 receptor expression (Sampson et al., 2008; Hilliard et al., 2011, 2013; Brown et al., 2012). It is therefore possible that signalling BJP via RXFP1–AT2 receptor heterodimers may contribute to the cardio-renal protection observed in young females and that this mechanism may be down-regulated post-menopause since the enhanced role for the AT2 receptors in females is lost with ageing and reproductive senescence (Mirabito et al., 2014a,b). This possibility warrants further investigation, as restoration of AT2 receptor expression in older women, or up-regulation in males, may represent a therapeutic target worth pursuing. In contrast, it is also possible that a deficit in relaxin may play a role in the progression of renal disease in females. In general, females are protected from renal disease except in the case of diabetic nephropathy, in which progression and severity of renal damage is greater in females (DiamondStanic et al., 2012; Barrett-Connor, 2013). Evidence suggests that this is due to a reduction in the oestrogen to testosterone ratio in females in response to the diabetic environment and a similar change in hormonal balance also occurs postmenopause, a time when the risk of cardio-renal disease starts to increase in women (Maric, 2009; Diamond-Stanic et al., 2012). Given the association between relaxin secretion and oestrogen status (Lippert et al., 1996; Seeger et al., 2000), relaxin may be altered in diabetic patients. However, rhRLX infusion in type I diabetic models has not been shown to improve renal fibrosis, for the reasons outlined above (Wong et al., 2013; Dschietzig et al., 2015). By contrast, in a high-fat diet model of insulin resistance, rhRLX infusion reversed collagen accumulation in the heart (Bonner et al., 2013). As these studies were all conducted in males, in the future, it will be important to examine the effect of rhRLX replacement therapy for diabetic nephropathy in females, in which an aged-related reduction in relaxin might be associated with sex hormone imbalance. Thus, several lines of evidence suggest that relaxin may explain the protection from cardiovascular and renal disease enjoyed by pre-menopausal women, while its lack may contribute to the exacerbation of disease in females. Anti-fibrotic effects of relaxin in the lung Fibrosis in the lung is seen histopathologically in asthma, chronic obstructive pulmonary disease, pulmonary fibrosis (including idiopathic pulmonary fibrosis), bronchopulmonary dysplasia and pulmonary hypertension. Its significance in these diseases is that it is associated with progression, severity of disease and resistance to treatment, and contributes to the functional end points of dyspnoea, airway hyperresponsiveness (AHR) and forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1). In a murine model of chronic allergic airways disease (AAD), which mimics several features of human asthma, systemic treatment with 0.5 mg·mL 1 rhRLX via mini-osmotic pumps was able to reduce total lung collagen and peribronchial ECM deposition to that seen in control mice (Royce et al., 2009). Furthermore, rhRLX treatment reversed epithelial thickening and significantly improved AHR but did not influence airway inflammation or goblet cell metaplasia. rhRLX therapy was also attempted in the most widely used preclinical model of parenchymal pulmonary fibrosis British Journal of Pharmacology (2017) 174 962–976 967 BJP C S Samuel et al. induced by bleomycin administration. In this model, mice are typically given a single or double dose of bleomycin – originally used for the treatment of cancers where fibrotic side effects in the lung were identified (Della Latta et al., 2015). Furthermore, rhRLX administration significantly reversed established parenchymal fibrosis as well myofibroblast contractility (Unemori et al., 1996; Pini et al., 2010; Huang et al., 2011). Given the fact that relaxin is a hormone and the RXFP1 receptor appears to be expressed widely throughout the body, it is important to limit off target effects. This is achievable for lung diseases as the organ is available for inhaled delivery. However, there are a number of issues with regard to lung delivery that must be addressed. In a murine AAD model of asthma, a similar reversal of established airway fibrosis was achieved with daily intranasal administration of rhRLX over 2 weeks (Royce et al., 2014a) as that observed in earlier studies in response to continuous systemic administration (Royce et al., 2009). Intranasal administration to mice involves simply allowing the mouse to inhale a bolus of rhRLX micropipetted to the nares. In humans, the lung is orders of magnitude larger, and hence, the challenge is for drugs inhaled at the nose or mouth to reach the small airways (<2 mm), which are of particular pathological significance (Thien, 2013), and the alveolar sacs. As such inhaled medications need to be delivered as dry powder or nebulized to achieve a small enough particle size to reach the terminal parts of the respiratory tree, rhRLX has yet to meet this, but promising new nebulization and delivery technologies are under development that enable a larger range of drugs to be nebulized without conformational changes or loss of bioactivity (Cortez-Jugo et al., 2015). It is well-documented that rhRLX has a strong antifibrotic effect especially against established fibrosis, particularly with its ability to down-regulate TGF-β1-mediated collagen production (Royce et al., 2014a; Unemori et al., 1996) and up-regulate gelatinases (MMP-2 and MMP-9) to help digest aberrant collagen accumulation in the lung and airway wall (Royce et al., 2009; Chow et al., 2012) (Figure 1). However, if rhRLX is to have potential as a treatment for fibrotic lung diseases, it is important that it is able to complement existing therapies, in particular inhaled and oral corticosteroids. rhRLX has been combined with methylprednisolone to treat established fibrosis in an experimental model of chronic AAD and it was found that combination therapy with the corticosteroid and rhRLX more effectively reduced subepithelial collagen thickness compared with either therapy alone (Royce et al., 2013). However, corticosteroids are not without their limitations, such as steroid resistance in some patients, side effects that limit optimal dosing in very young children and, from the point of view of fibrosis, the fact that blocking inflammation does not eliminate fibrosis, as is clear in the adult asthma population where airway remodelling remains rife despite corticosteroids being in widespread use for decades. This reflects the fact that airway remodelling and fibrosis are often not due to chronic inflammation alone but arise from other aetiologies such as epithelial damage and genetic susceptibility (Holgate et al., 2006). MSCs are another therapeutic approach used experimentally in combination with rhRLX for lung disease treatment (Royce et al., 2015). Used alone, bone marrow-derived MSCs 968 British Journal of Pharmacology (2017) 174 962–976 have been shown to have potential for treating a range of fibrotic lung diseases (Royce et al., 2014b) and have been used successfully to treat AAD (Ogulur et al., 2014) and bleomycin-induced (Ortiz et al., 2003; Moodley et al., 2009) disease models. Stem cell therapies, in general, are thought to heal either by engraftment into the host tissue or by secretion of exosomes and proteins that have paracrine and immunomodulatory effects in diseased tissue. In many inflammatory lung diseases, there are areas of epithelial damage where engraftment of MSCs would conceivably be of benefit, but it seems more plausible that secretions of MSCs explain most of the beneficial effects seen in mouse models (Ge et al., 2013). It was hypothesized that combining rhRLX with MSCs would augment the therapeutic efficacy of the latter, by improving the micro-environment via removal of aberrant ECM accumulation and fibrotic stimuli (Formigli et al., 2007; Huuskes et al., 2015). In a chronic AAD model, this combination treatment further reversed disease-induced airway inflammation and airway/lung fibrosis compared with either treatment alone and further increased MMP-2 and MMP-9 levels (Royce et al., 2015). Another avenue for this method is for the overexpression of relaxin by MSCs, which would provide both a novel delivery method of rhRLX and the likely positive effects of rhRLX in the immediate microenvironment of the MSC. This protein overexpression has already been used with other MSC treatments including overexpression of the CXCR4 receptor to enhance homing (Yang et al., 2015) and with murine C2C12 myoblasts overexpressing relaxin to reduce fibrosis and promote angiogenesis in ischaemically damaged organs (Formigli et al., 2007). Much progress has been made on rhRLX in the acute heart failure trials towards being a realistic drug treatment for human disease. In addition, it has a good safety profile, the ability to reverse aberrant collagen (without effecting basal ECM required for normal structure) and other advantages compared with other anti-fibrotic drugs both currently used and under development. Given these factors and the outstanding need for novel therapies to treat lung diseases such as asthma (estimated 235 million sufferers 2004) and chronic obstructive pulmonary disease (64 million sufferers) (World Health Organization Global Burden of Disease), relaxin has great potential as a future therapy for fibrosis associated with various lung diseases. Anti-fibrotic effects of relaxin in the liver Hepatic fibrosis results from the accumulation of fibrillar collagens and other ECM proteins in response to chronic injury from a variety of causes, including alcohol overuse, viral hepatitis and nonalcoholic steatohepatitis (Trautwein et al., 2015). The liver cells responsible for the production of the majority of the collagen in hepatic fibrosis are the hepatic stellate cells (HSCs), specialized cells that reside in the perisinusoidal regions (Friedman, 2008; Puche et al., 2013). HSCs are unusual in that, in the normal (quiescent) state, they function as lipid storing cells and are not fibroblastic in nature. With liver injury, inflammatory cytokines such as TGF-β1 and PDGF stimulate HSCs, causing their transdifferentiation to an activated myofibroblastic Anti-fibrotic actions of relaxin phenotype, characterized by proliferation, contractility and high expression of α-smooth muscle actin (α-SMA), and fibrillar collagens. At the same time, HSCs secrete TIMP-1 and TIMP-2 (Friedman, 2008; Puche et al., 2013), resulting in a shift in the balance between matrix production and degradation and a net increase in collagen accumulation. When the cause of liver injury is removed, the number of activated HSCs is decreased, through either apoptosis or reversion to the quiescent phenotype, allowing clearance of the excess collagen and a return to the normal liver architecture and functioning. With chronic injury, however, the collagen accumulation causes disruption of liver architecture and can eventually lead to cirrhosis. For this reason, HSCs have been a prime target in the development of new treatments for advanced hepatic fibrosis and cirrhosis (Trautwein et al., 2015). The first study of relaxin’s effects in the liver was in rats treated for 4 days with porcine relaxin (Bani et al., 2001). Relaxin caused dilation of the sinusoids and changes in contractile, actin-rich cells that were probably either myofibroblasts or activated HSCs. The HSCs were confirmed as a target of relaxin in studies using culture-activated rat HSCs, where rhRLX caused a decreased collagen deposition and synthesis, accompanied by decreased secretion of TIMP-1 and TIMP-2, but without a change in the gene expression of MMPs or α-SMA (Williams et al., 2001). In another study, porcine relaxin treatment of culture-activated rat HSCs also caused decreased collagen synthesis and deposition, as well as decreased secretion of type I collagen, TIMP-1 and TIMP-2 (Bennett et al., 2003). Furthermore, relaxin decreased the level of α-SMA protein, increased the degradation of type I collagen and increased the expression of MMP-13, the major rodent fibrillar collagenase. Similar results were observed using primary human HSCs (Fallowfield et al., 2014). Taken together, these studies provided evidence that human or porcine relaxin can inhibit the pro-fibrotic properties of activated HSC and promote conditions that favour collagen degradation (Figure 1). HSCs are the major liver cells that express RXFP1. Early studies using PCR detected RXFP1 expression in human liver (Hsu et al., 2002; Hsu et al., 2003), but not female mouse liver (Scott et al., 2006). It was later revealed that quiescent rat HSCs express very low levels of the RXFP1 receptor, which increased markedly during transdifferentiation in culture, and the RXFP1 receptor was detected in fibrotic mouse liver and cirrhotic human liver (Bennett et al., 2005, 2007). These findings were later confirmed in primary rat and human HSCs and in rat and human hepatic fibrosis (Fallowfield et al., 2014). The signalling pathways activated by relaxin in HSCs are consistent with those in fibroblasts from other tissues and include cAMP, cGMP, NO and Akt (Bennett et al., 2007; Fallowfield et al., 2014). Furthermore, recent findings suggest that rhRLX activation of RXFP1 receptors can result in activation of the anti-fibrotic transcription factor PPARγ, through a mechanism involving cAMP, PKA, p38-MAPK and PPARγ coactivator protein 1α (PGC1α; Singh and Bennett, 2010; Singh et al., 2015). Several studies have been conducted to assess the efficacy of relaxin in the treatment of hepatic fibrosis. The first was a prevention study using CCl4-induced hepatic fibrosis in rats, with concomitant treatment with rhRLX (~0.5 mg·kg 1·day 1) for 4 weeks (Williams et al., 2001), in which rhRLX reduced BJP the overall liver collagen content as assessed by hydroxyproline content. Later studies focused on more clinically relevant models of established hepatic fibrosis in mice, using 4 weeks of CCl4 treatment to induce fibrosis, followed by rhRLX treatment via osmotic pumps with continued CCl4 administration. In one study using short-term treatment with porcine relaxin (0.5 mg·kg 1·day 1), relaxin had a small effect on fibrosis after 1 week, but little effect after 2 weeks (Bennett et al., 2009). A later study of 4 week treatment with rhRLX (25 or 75 μg·kg 1·day 1) showed significant reductions in the levels of liver collagen and α-SMA and reduced expression of types I and III collagen, α-SMA and TIMP-2, increased expression of MMP-3 and MMP-13 and increased collagen degrading activity (Bennett et al., 2014). Finally, another study showed that with short-term (72 h) administration of 0.5 mg·kg 1·day 1 rhRLX into rats made fibrotic with 8 weeks of CCl4, rhRLX reduced the amount of α-SMA protein, but not the amount of liver collagen, and decreased the gene expression of type I collagen, α-SMA, MMP-3 and TGF-β (Fallowfield et al., 2014). Furthermore, in both CCl4 and bile duct ligation models, rhRLX reduced portal pressure and increased the hepatic oxygen supply, consistent with other studies using perfused liver models (Boehnert et al., 2008; Fallowfield et al., 2014). Taken together, the studies conducted thus far show some positive effects of relaxin on hepatic fibrosis and haemodynamics, at least in part by reducing the activated phenotype of HSCs and/or promoting matrix degradation. However, there are some differences in the various experimental models used thus far, and further studies are needed to determine the optimal conditions necessary for effective relaxin treatment of hepatic fibrosis. Nevertheless, these collective preclinical results led to a phase II trial of recombinant human relaxin on haemodynamics in patients with compensated cirrhosis and portal hypertension, which was completed in early 2015 (clinicaltrials.gov identifier NCT01640964). The results of this trial, when released, may clarify the potential role of relaxin in the treatment of human liver disease. Anti-fibrotic effects of relaxin on the integumentary system and connective tissues Research involving relaxin’s effect on clinical skin pathology has a notable past, with scleroderma as the most frequently studied pathology. Scleroderma is a spectrum disorder and includes a milder, limited form as well as a ‘diffuse’ form and involves fibrosis of internal organs. In its limited form, common manifestations in the skin are limited to the extremities and include skin tightness and ulcerations, thickening, altered vascularity and sensitivity of the hands and digits (Raynaud’s syndrome), difficulty in swallowing (dysphagia) and localized dilation of small blood vessels (CREST syndrome). In diffuse scleroderma, major organ fibrosis occurs along with widespread skin involvement. Early reports indicated that oestrogen priming followed by 20–40 mg doses of porcine relaxin (then known as ‘releasin’) partially resolved skin tightness and Raynaud’s symptoms, prompted healing of skin ulcerations that were refractory to corticosteroid British Journal of Pharmacology (2017) 174 962–976 969 BJP C S Samuel et al. treatment and, in limited cases, relieved dysphagia (Casten and Boucek, 1958; Ismay, 1958; Evans, 1959; Rivelis et al. 1965). No improvements were reported in heart, lung or kidney function. Therapeutic effectiveness often required weeks of therapy (3–5 weeks; Casten and Boucek, 1958) with therapy continuing for protracted periods (up to 30 months), with discontinuation often exacerbating symptoms. Reports of complications were rare, mainly focused on injection site soreness. Contrary to these reports, others observed treatment failure in four patients treated with 80 mg of releasin for 3–4 weeks (Jefferis and Dixon, 1962). These reports suggested that treatment efficacy was most often observed with continued therapy and that response variability was present in the outcomes. More recently, clinical safety trials were conducted to establish tolerability of rhRLX to treat patients with systemic disease of short (<5 year) duration. A 24 week randomized control trial showed both safety and preliminary measures of efficacy for 25 μg·kg 1·day 1 subcutaneous rhRLX infusion; a higher dose (100 μg·kg 1·day 1), however, failed to show an advantage over the placebo (Seibold et al., 2000). A 14% improvement over placebo in ratings of standardized scoring of skin thickness was observed in this trial. A subsequent FDA phase III efficacy randomized clinical trial involving over 200 patients failed to show efficacy over placebo and evidenced seven instances of serious adverse events involving renal crisis (Khanna et al., 2009). The failure of this phase III trial forced a reassessment of relaxin therapeutics for treating skin-associated fibrosis, most especially in the context of targeting tissue competence to respond to relaxin. Certainly, in vitro studies have lent support for therapeutic trials (Unemori and Amento, 1990). rhRLX (1–100 ng·mL 1) dose-dependently reduced type I collagen production from most (but not all) of a small sample of biopsies derived from patients with scleroderma (Unemori et al., 1992), and these effects were synergistically enhanced by co-administration of IFN-γ (100 U·mL 1). Furthermore, rhRLX significantly improved the cosmetic appearance and histology of porcine excisional wounds when administered at 130 μg·kg 1·day 1 for 6 weeks (Stewart, 2009). Currently, studies are underway to determine the effect of relaxin treatment in animal models of dermal wounding, and the expression of relaxin receptors in biopsy samples from patients with Dupuytren’s disease (Cooney, unpublished data). That fibroblasts are likely targets for relaxin therapy is also supported from other clinically relevant anatomical sites, suggesting a systemic-wide tropism. Treatment with rhRLX (up to 100 ng·mL 1) of primary cultures of human fibroblasts obtained from vaginal wall biopsies resulted in reduced TGF-β 1 translation (Wen et al., 2008). Fibroblasts comprising knee ligaments and thumb express relaxin receptors (Dragoo et al., 2003; Galey et al., 2003; Lubahn et al., 2006). An in situ study of female lower shank tendons showed an inverse relationship between serum relaxin levels and patellar tendon stiffness (Pearson, et al., 2011). These data strongly suggest that fibroblasts located in connective tissue are competent to respond to rhRLX. However, the scope of relaxin’s apparent cellular interaction within the integument extends beyond resident fibroblasts. Several studies have shown that keratinocytes derived from normal skin express RXFP1 receptors (Cooney 970 British Journal of Pharmacology (2017) 174 962–976 et al., 2010; Giordano et al., 2012). Relaxin effects have also been isolated to a substituent of the ECM, fibrillin-2, in the form of down-regulated gene transcription and translation in human dermal fibroblasts in response to rhRLX dosing at 30 ng·mL 1 (Samuel et al., 2003b). Cell lines derived from the eye epithelium and surrounding sites (human cornea, conjunctiva, sebaceous and lacrimal glands) express both RXFP1 and RXFP2 receptors and respond to dosing with INSL3 and rhRLX (Hampel et al., 2012; Hampel et al., 2013). Blood vessel endothelium and endothelial precursors have been shown to be rhRLX targets. The administration of rhRLX 0.1 mg·kg 1·day 1, s.c., to rats enhanced angiogenesis in wound chambers, increased VEGF and bFGF signalling and production and ultimately determined that effects were likely to be mediated by monocytes infiltrating hypoxic tissue areas (Unemori et al., 2000). Importantly, effects were confined to the site of wounding and were not seen in alveolar pulmonary macrophages. Others showed increases in the transcription and translation of mediators of angiogenesis and vasculogenesis in genetically diabetic mice dosed with 25 μg·day 1 of porcine relaxin with consequent improvement in wound healing time (Bitto et al., 2013) and that rhRLX signalled through RXFP1 receptors to induce mobilization of bone marrow-derived endothelial progenitors (Segal et al., 2012). Overall, there are an abundance of histological sites in the integument that express relaxin receptors and respond to dosing in vitro. Limitations of using rhRLX as a therapeutic and challenges to consider for future studies Whilst these collective findings highlight the therapeutic potential of rhRLX as an anti-fibrotic, there are a number of recognized barriers to clinical translation that are noteworthy. Firstly, the clinical efficacy of various anti-fibrotic agents is difficult to evaluate in general due to the lack of reliable biomarkers that could indicate when fibrosis has developed to the point when these agents should be appropriately administered, the lack of reliable non-invasive end points that could demonstrate if they have regressed disease progression, and the slow pathogenesis of fibrosis in humans (compared to that in animals), which can take decades to develop. Secondly, rhRLX is a peptide-based drug with a short in vivo half-life (~10 min; Nair et al., 2015). Although this has been overcome at the experimental level by administering rhRLX to various animal models via subcutaneously implanted osmotic mini-pumps and at the clinical level via microinfusion pumps (Erikson and Unemori, 2001), weekly injections (McGorray et al., 2012) or intravenous administration (Weiss et al., 2009), the relatively short duration of rhRLX treatment may have contributed to the failed clinical trials to date that have attempted to demonstrate its antifibrotic efficacy in patients with varying conditions, particularly given that fibrosis is a chronic condition that is likely to require long-term treatment to diminish its progression. Thirdly, the requirement for longer term rhRLX treatment will only add to its expense to produce. While small molecules that activate RXFP1 receptors (Xiao et al., 2013) and single-chain derivatives of rhRLX (Hossain et al., 2011b; Hossain et al., 2016) have recently been developed and will be cheaper to manufacture, further work is required to Anti-fibrotic actions of relaxin determine their anti-fibrotic efficacy when chronically administered over long periods. The latter might be difficult to test in rodent models though which have been found to mount antibody responses to rhRLX, which in turn have caused increased and variable circulating levels of rhRLX beyond 10 days post-administration (personal communication; Dr. Elaine Unemori; Corthera Inc. and Novartis AG, San Mateo, CA, USA). Consistent with this, mice administered 0.5 mg·kg 1·day 1 rhRLX undergo a doubling of circulating human relaxin levels after 14 days of administration compared with that after 5 days (Samuel et al., 2003a), which may lead to higher concentrations of the drug producing lower physiological responses, given its well-demonstrated bell-shaped dose–response effects (Unemori et al., 1996; Danielson and Conrad, 2003; Teerlink et al., 2009). Furthermore, the importance of understanding tissue competence to respond to rhRLX along with signalling pathways is paramount to ensuring both efficacy and safety of rhRLX in human clinical trials. Recently, Giordano et al. (2012 showed that skin biopsies from patients with limited systemic scleroderma showed weak or no staining for RXFP1 receptors. In line with this, only 30% of Dupuytren’s nodules stain positive for the RXFP1 receptor (Cooney, unpublished observation). These findings beg the question of how receptor expression changes over the course of disease. Restoration of calcium signalling via NO is impaired in the endothelium of spontaneously hypertensive rats and cannot be remediated by rhRLX, unlike that of Wistar Kyoto control rats (Nistri et al., 2015), suggesting differential responsiveness based on phenotype or pathological state. Indeed, as in the disease state, alterations in the ECM environment of cell culture matter, especially in the context of myofibroblasts. Material properties of the ECM affect mechanical signal transduction in myofibroblasts, which, in turn, alters gene transcription (Vi et al., 2010). These data show that culturing myofibroblasts on tissue culture plastic or even type I collagen substrates significantly alters the feedback loop between cells and ECM and underscores the importance of recognizing the distinction between the pathological environment versus one that fosters cell growth and proliferation. Conclusions rhRLX continues to hold promise as a therapeutic to treat several fibrotic conditions associated with up-regulated TGF-β1 and myofibroblast burden. In addition to directly downregulating intracellular Smad activity to suppress TGF-β1 signal transduction and the pro-fibrotic actions of TGF-β1 on myofibroblast differentiation and aberrant ECM synthesis, it is able to regulate matrix degradation by altering the MMP-to-TIMP balance (summarized in Figure 1). Additionally, it may also indirectly inhibit fibrosis progression via its other organ-protective actions, including its anti-inflammatory, anti-hypertrophic, anti-apoptotic, angiogenic and vasodilatory effects, and can also promote wound healing and tissue function through these combined actions. Although a number of high-quality randomized trials to date have failed to demonstrate the clinical efficacy of its matrix remodelling actions in human subjects (which have been well characterized in various experimental models), it is BJP possible that the positive effects of rhRLX on blood pressure and renal function that have been observed in various trials may be associated with its ability to inhibit fibrosis progression in affected organs. A significant challenge for future trials is to identify patients who may benefit from rhRLX therapy, identify appropriate end points for a peptide-based therapeutic, which requires continuous administration, consider the timing and length of treatment as well as appropriate route of administration and ensure tissue penetrability beyond the direct site of administration. Application of gene sequencing to human biopsy material (to elucidate whether perturbations involving single nucleotide polymorphisms or deletions/additions that impair RXFP1 receptor functioning affect therapeutic responses to rhRLX), and the development of small molecule mimetics of relaxin that have improved pharmacokinetics and biodistribution properties compared with native H2 relaxin, will undoubtedly aid this effort. Cell culture models that better mimic in situ tissue conditions will also provide more relevant information. Overall, this field of relaxin investigation remains vibrant and full of potential. Acknowledgements This article was supported in part by National Health & Medical Research Council (NHMRC) of Australia Senior Research Fellowships to C.S.S. (GNT1041766) and K.M.D. (GNT1041844) and NHMRC Project Grants to T.D.H. and C. S.S. (GNT1078694) and to K.M.D., C.S.S and T.D.H. (GNT1101552) and by US Department of Veterans Affairs Merit Review (BX000849) and National Institutes of Health (AA015509) Grants to R.G.B. Conflict of interest The authors declare no conflicts of interest. References Alexander SP, Cidlowski JA, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015c). The concise guide to pharmacology 2015/16: nuclear hormone receptors. Br J Pharmacol 172: 5956–5978. Alexander SP, Davenport AP, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015b). The concise guide to pharmacology 2015/16: G protein-coupled receptors. Br J Pharmacol 172: 5744–5869. Alexander SP, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015d). The concise guide to pharmacology 2015/16: enzymes. Br J Pharmacol 172: 6024–6109. Alexander SP, Kelly E, Marrion N, Peters JA, Benson HE, Faccenda E et al. (2015a). The concise guide to pharmacology 2015/16: overview. Br J Pharmacol 172: 5279–5243. Bani D (2008). Relaxin as a natural agent for vascular health. Vasc Health Risk Manage 4: 515–524. Bani D, Nistri S, Quattrone S, Bigazzi M, Bani Sacchi T (2001). The vasorelaxant hormone relaxin induces changes in liver sinusoid British Journal of Pharmacology (2017) 174 962–976 971 BJP C S Samuel et al. microcirculation: a morphologic study in the rat. J Endocrinol 171: 541–549. Barrett-Connor E (2013). Gender differences and disparities in allcause and coronary heart disease mortality: epidemiological aspects. Best Pract Res Clin Endocrinol Metab 27: 481–500. Cooney TE, Gor RA, Pfaff DW, Schober JM (2010). Detection of RXFP1 receptors in skin biopsies from children with congenital adrenal hyperplasia: a preliminary report. J Pediatr Urol 6: 389–395. Conrad KP (2004). Mechanisms of renal vasodilation and hyperfiltration during pregnancy. J Soc Gynecol Investig 11: 438–448. Bathgate RA, Ivell R, Sanborn BM, Sherwood OD, Summers RJ (2006). International Union of Pharmacology LVII: recommendations for the nomenclature of receptors for relaxin family peptides. Conrad KP, Debrah DO, Novak J, Danielson LA, Shroff SG (2004). Relaxin modifies systemic arterial resistance and compliance in conscious, nonpregnant rats. Endocrinology 145: 3289–3296. Bathgate RA, Lekgabe ED, McGuane JT, Su Y, Pham T, Ferraro T et al. (2008). Adenovirus-mediated delivery of relaxin reverses cardiac fibrosis. Mol Cell Endocrinol 280: 30–38. Conrad KP, Shroff SG (2011). Effects of relaxin on arterial dilation, remodeling, and mechanical properties. Curr Hypertens Rep 13: 409–420. Bennett RG, Dalton SR, Mahan KJ, Gentry-Nielsen MJ, Hamel FG, Tuma DJ (2007). Relaxin receptors in hepatic stellate cells and cirrhotic liver. Biochem Pharmacol 73: 1033–1040. Cortez-Jugo C, Qi A, Rajapaksa A, Friend JR, Yeo LY (2015). Pulmonary monoclonal antibody delivery via a portable microfluidic nebulization platform. Biomicrofluidics 9: 052603. Bennett RG, Heimann DG, Tuma DJ (2009). Relaxin reduces fibrosis in models of progressive and established hepatic fibrosis. Ann N Y Acad Sci 1160: 348–349. Danielson LA, Conrad KP (2003). Time course and dose response of relaxin-mediated renal vasodilation, hyperfiltration, and changes in plasma osmolality in conscious rats. J Apply Physiol 95: 1509–1514. Bennett RG, Kharbanda KK, Tuma DJ (2003). Inhibition of markers of hepatic stellate cell activation by the hormone relaxin. Biochem Pharmacol 66: 867–874. Danielson LA, Kercher LJ, Conrad KP (2000). Impact of gender and endothelin on renal vasodilation and hyperfiltration induced by relaxin in conscious rats. Am J Physiol Regul Integr Comp Physiol 279: R1298–R1304. Bennett RG, Mahan KJ, Gentry-Nielsen MJ, Tuma DJ (2005). Relaxin receptor expression in hepatic stellate cells and in cirrhotic rat liver tissue. Ann N Y Acad Sci 1041: 185–189. Bennett RG, Heimann DG, Singh S, Simpson RL, Tuma DJ (2014). Relaxin decreases the severity of established hepatic fibrosis in mice. Liver Int 34: 416–426. Bitto A, Irrera N, Minutoli L, Calò M, Lo Cascio P, Caccia P et al. (2013). Relaxin improves multiple markers of wound healing and ameliorates the disturbed healing pattern of genetically diabetic mice. Clin Sci 125: 575–585. Boehnert MU, Armbruster FP, Hilbig H (2008). Relaxin as a protective substance in preservation solutions for organ transplantation, as shown in an isolated perfused rat liver model. Transplant Proc 40: 978–980. Bonacchi M, Nistri S, Nanni C, Gelsomino S, Pini A, Cinci L et al. (2009). Functional and histopathological improvement of the postinfarcted rat heart upon myoblast cell grafting and relaxin therapy. J Cell Mol Med 13: 3437–3448. Bonner JS, Lantier L, Hocking KM, Kang L, Owolabi M, James FD et al. (2013). Relaxin treatment reverses insulin resistance in mice fed a high-fat diet. Diabetes 62: 3251–3260. Brown RD, Hilliard LM, Head GA, Jones ES, Widdop RE, Denton KM (2012). Sex differences in the pressor and tubuloglomerular feedback response to angiotensin II. Hypertension 59: 129–135. Casten GG, Boucek RJ (1958). Use of relaxin to treat scleroderma. JAMA 166: 319–324. Della Latta V, Cecchettini A, Del Ry S, Morales MA (2015). Bleomycin in the setting of lung fibrosis induction: from biological mechanisms to counteractions. Pharmacol Res 97: 122–130. Diamond-Stanic MK, You YH, Sharma K (2012). Sugar, sex, and TGFbeta in diabetic nephropathy. Semin Nephrol 32: 261–268. Dragoo JL, Lee RS, Benhaim P, Finerman GA, Hame SL (2003). Relaxin receptors in the human female anterior cruciate ligament. Am J Sports Med 31: 577–584. Dschietzig T, Bartsch C, Kinkel T, Baumann G, Stangl K (2005). Myocardial relaxin counteracts hypertrophy in hypertensive rats. Ann N Y Acad Sci 1041: 441–443. Dschietzig TB, Krause-Relle K, Hennequin M, von Websky K, Rahnenfuhrer J, Ruppert J et al. (2015). Relaxin-2 does not ameliorate nephropathy in an experimental model of type-1 diabetes. Kidney Blood Press Res 40: 77–88. Du XJ, Bathgate RA, Samuel CS, Dart AM, Summers RJ (2010). Cardiovascular effects of relaxin: from basic science to clinical therapy. Nature Revs Cardiology 7: 48–58. Du XJ, Samuel CS, Gao XM, Zhao L, Parry LJ, Tregear GW (2003). Increased myocardial collagen and ventricular diastolic dysfunction in relaxin deficient mice: a gender-specific phenotype. Cardiovasc Res 57: 395–404. Erikson MS, Unemori EN (2001). Relaxin clinical trials in systemic sclerosis. In: Tregear GW, Ivell R, Bathgate RA, Wade JD (eds). Relaxin 2000: Proceedings of the Third International Conference on Relaxin & Related Peptides. Kluwer Academic Publishers: The Netherlands, pp. 373–381. Chan LJ, Rosengren KJ, Layfield SL, Bathgate RA, Separovic F, Samuel CS et al. (2012). Identification of key residues essential for the structural fold and receptor selectivity within the A-chain of human gene-2 (H2) relaxin. J Biol Chem 287: 41152–41164. Evans JA (1959). Relaxin (releasin) therapy in diffuse progressive scleroderma; a preliminary report. AMA Arch Derm 79: 150–158. Chow BS, Chew EG, Zhao C, Bathgate RA, Hewitson TD, Samuel CS (2012). Relaxin signals through a RXFP1–pERK–nNOS–NO–cGMPdependent pathway to up-regulate matrix metalloproteinases: the additional involvement of iNOS. PLoS One 7 .e42714 Fallowfield JA, Hayden AL, Snowdon VK, Aucott RL, Stutchfield BM, Mole DJ et al. (2014). Relaxin modulates human and rat hepatic myofibroblast function and ameliorates portal hypertension in vivo. Hepatology 59: 1492–1504. Chow BS, Kocan M, Bosnyak S, Sarwar M, Wigg B, Jones ES et al. (2014). Relaxin requires the angiotensin II type 2 receptor to abrogate renal interstitial fibrosis. Kidney Int 86: 75–85. Formigli L, Perna AM, Meacci E, Cinci L, Margheri M, Nistri S et al. (2007). Paracrine effects of transplanted myoblasts and relaxin on post-infarction heart remodelling. J Cell Mol Med 11: 1087–1100. 972 British Journal of Pharmacology (2017) 174 962–976 Anti-fibrotic actions of relaxin Friedman SL (2008). Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev 88: 125–172. Frieler RA, Mortensen RM (2015). Immune cell and other noncardiomyocyte regulation of cardiac hypertrophy and remodeling. Circulation 131: 1019–1030. Galey S, Konieczko EM, Arnold CA, Cooney TE (2003). Immunohistological detection of relaxin binding to anterior cruciate ligaments. Orthopedics 26: 1201–1204. Garber SL, Mirochnik Y, Brecklin C, Slobodskoy L, Arruda JA, Dunea G (2003). Effect of relaxin in two models of renal mass reduction. Am J Nephrol 23: 8–12. Garber SL, Mirochnik Y, Brecklin CS, Unemori EN, Singh AK, Slobodskoy L et al. (2001). Relaxin decreases renal interstitial fibrosis and slows progression of renal disease. Kidney Int 59: 876–882. Ge X, Bai C, Yang J, Lou G, Li Q, Chen R (2013). Effect of mesenchymal stem cells on inhibiting airway remodeling and airway inflammation in chronic asthma. J Cell Biochem 114: 1595–1605. Giordano N, Volpi N, Franci D, Corallo C, Fioravanti A, Papakostas P et al. (2012). Expression of RXFP1 in skin of scleroderma patients and control subjects. Scand J Rheumatol 41: 391–395. BJP Holgate ST, Holloway J, Wilson S, Howarth PH, Haitchi HM, Babu S et al. (2006). Understanding the pathophysiology of severe asthma to generate new therapeutic opportunities. J Allergy Clin Immunol 117: 496–506. Hossain MA, Kocan M, Yao ST, Royce SG, Nair VB, Siwek C et al. (2016). A single-chain derivative of the relaxin hormone is a functionally selective agonist of the G protein-coupled receptor, RXFP1. Chem Sci . doi:10.1039/C5SC04754. Hossain MA, Man BC, Zhao C, Xu Q, Du XJ, Wade JD et al. (2011a). H3 relaxin demonstrates antifibrotic properties via the RXFP1 receptor. Biochemistry 50: 1368–1375. Hossain MA, Rosengren KJ, Samuel CS, Shabanpoor F, Chan LJ, Bathgate RA et al. (2011b). The minial active structure of human relaxin-2. J Biol Chem 286: 37555–37565. Hsu SY, Nakabayashi K, Nishi S, Kumagai J, Kudo M, Bathgate RA et al. (2003). Relaxin signalling in reproductive tissues. Mol Cell Endocrinol 202: 165–170. Hsu SY, Nakabayashi K, Nishi S, Kumagai J, Kudo M, Sherwood OD et al. (2002). Activation of orphan receptors by the hormone relaxin. Science 295: 671–674. Gu HP, Lin S, Xu M, Yu HY, Du XJ, Zhang YYet al. (2012). Upregulating relaxin expression by G-quadruplex interactive ligand to achieve antifibrotic action. Endocrinology 153: 3692–3700. Huang X, Gai Y, Yang N, Lu B, Samuel CS, Thannickal VJ et al. (2011). Relaxin regulates myofibroblast contractility and protects against lung fibrosis. Am J Path 179: 2751–2765. Hampel U, Klonisch T, Makrantonaki E, Sel S, Schulze U, Garreis F et al. (2012). Relaxin 2 is functional at the ocular surface and promotes corneal wound healing. Invest Ophthalmol Vis Sci 53: 7780–7790. Huuskes BM, Wise AF, Cox AJ, Lim EX, Payne NL, Kelly DJ et al. (2015). Combination therapy of mesenchymal stem cells and serelaxin effectively attenuates renal fibrosis in obstructive nephropathy. FASEB J 29: 540–553. Hampel U, Klonisch T, Sel S, Schulze U, Garreis F, Seitmann H et al. (2013). Insulin-like factor 3 promotes wound healing at the ocular surface. Endocrinology 154: 2034–2045. Ismay G (1958). Relaxin: its effects in a case of acrosclerosis. Br J Dermatol 70: 171–175. Heeg MHJ, Koziolek MJ, Vasko R, Schaefer L, Sharma K, Muller GA et al. (2005). The antifibrotic effects of relaxin in human renal fibroblasts are mediated in part by inhibition of the Smad2 pathway. Kidney Int 68: 96–109. Henry B, Gabris B, Li Q, Martin B, Giannini M, Parikh A et al. (2016). Relaxin suppresses atrial fibrillation in aged rats by reversing fibrosis and upregulating Na-channels. Heart Rhythm 13: 983–991. Hewitson TD (2009). Renal tubulointerstitial fibrosis: common but never simple. Am J Physiol Renal Physiol 296: F1239–F1244. Hewitson TD, Ho WY, Samuel CS (2010). Antifibrotic properties of relaxin: in vivo mechanism of action in experimental renal tubulointerstitial fibrosis. Endocrinology 151: 4938–4948. Hewitson TD, Holt SG, Smith ER (2015). Animal models to study links between cardiovascular disease and renal failure and their relevance to human pathology. Front Immunol 6: 465. Hewitson TD, Zhao C, Wigg B, Lee SW, Simpson ER, Boon WC et al. (2012). Relaxin and castration in male mice protect from, but testosterone exacerbates, age-related cardiac and renal fibrosis, whereas estrogens are an independent determinant of organ size. Endocrinology 153: 188–199. Hilliard LM, Sampson AK, Brown RD, Denton KM (2013). The “his and hers” of the renin–angiotensin system. Curr Hypertens Rep 15: 71–79. Hilliard LM, Nematbakhsh M, Kett MM, Teichman E, Sampson AK, Widdop RE et al. (2011). Gender differences in pressure-natriuresis and renal autoregulation: role of the Angiotensin type 2 receptor. Hypertension 57: 275–282. Jefferis JE, Dixon AS (1962). Failure of relaxin in the treatment of scleroderma. Ann Rheum Dis 21: 295–297. Jeyabalan A, Novak J, Danielson LA, Kerchner LJ, Opett SL, Conrad KP (2003). Essential role for vascular gelatinase activity in relaxininduced renal vasodilation, hyperfiltration, and reduced myogenic reactivity of small arteries. Circ Res 93: 1249–1257. Khanna D, Clements PJ, Furst DE, Korn JH, Ellman M, Rothfield N et al. (2009). Relaxin Investigators and the Scleroderma Clinical Trials Consortium. Recombinant human relaxin in the treatment of systemic sclerosis with diffuse cutaneous involvement: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum 60: 1102–1111. Lekgabe ED, Kiriazis H, Zhao C, Xu Q, Moore XL, Su Yet al. (2005). Relaxin reverses cardiac and renal fibrosis in spontaneously hypertensive rats. Hypertension 46: 412–418. Lekgabe ED, Royce SG, Hewitson TD, Tang ML, Zhao C, Moore XL et al. (2006). The effects of relaxin and estrogen deficiency on collagen deposition and hypertrophy of non-reproductive organs. Endocrinology 147: 5575–5583. Ling LH, Kistler PM, Kalman JM, Schilling RJ, Hunter RJ (2016). Comorbidity of atrial fibrillation and heart failure. Nat Revs Cardiology 13: 131–147. Lippert TH, Armbruster FP, Seeger H, Mueck AO, Zwirner M, Voelter W (1996). Urinary excretion of relaxin after estradiol treatment of postmenopausal women. Clin Exp Obstet Gynecol 23: 65–69. Lubahn J, Ivance D, Konieczko E, Cooney T (2006). Immunohistochemical detection of relaxin binding to the volar oblique ligament. J Hand Surg Am 31: 80–84. British Journal of Pharmacology (2017) 174 962–976 973 BJP C S Samuel et al. Mandavia CH, Aroor AR, Demarco VG, Sowers JR (2013). Molecular and metabolic mechanisms of cardiac dysfunction in diabetes. Life Sci 92: 601–608. Masterson R, Hewitson TD, Kelynack K, Martic M, Parry L, Bathgate R et al. (2004). Relaxin down-regulates renal fibroblast function and promotes matrix remodelling in vitro. Nephrol Dial Transpl 19: 544–552. McDonald GA, Sarkar P, Rennke H, Unemori E, Kalluri R, Sukhatme VP (2003). Relaxin increases ubiquitin-dependent degradation of fibronectin in vitro and ameliorates renal fibrosis in vivo. Am J Physiol Renal Physiol 285: F59–F67. McGorray SP, Dolce C, Kramer S, Stewart D, Wheeler TT (2012). A randomized, placebo-controlled clinical trial on the effects of recombinant human relaxin on tooth movement and short-term stability. Am J Orthod Dentofacial Orthop 141: 196–203. McGuane JT, Danielson LA, Debrah JE, Rubin JP, Novak J, Conrad KP (2011a). Angiogenic growth factors are new and essential players in the sustained relaxin vasodilatory pathway in rodents and humans. Hypertension 57: 1151–1160. McGuane JT, Debrah JE, Sautina L, Jarajapu YP, Novak J, Rubin JP et al. (2011b). Relaxin induces rapid dilation of rodent small renal and human subcutaneous arteries via PI3 kinase and nitric oxide. Endocrinology 152: 2786–2796. Maric C (2009). Sex, diabetes and the kidney. Am J Physiol Renal Physiol 296: F680–F688. Metra M, Cotter G, Davison BA, Felker GM, Filippatos G, Greenberg BH et al. (2013). Effect of serelaxin on cardiac, renal, and hepatic biomarkers in the Relaxin in Acute Heart Failure (RELAX-AHF) development program: correlation with outcomes. J Am Coll Cardiol 61: 196–206. Mirabito K, Hilliard L, Head G, Widdop R, Denton K (2014a). Pressor responsiveness to angiotensin II in female mice is enhanced with age: role of the angiotensin type 2 receptor. Biol Sex Diff 5: 13. Mirabito KM, Hilliard LM, Kett MM, Brown RD, Booth SC, Widdop RE et al. (2014b). Sex- and age-related differences in the chronic pressure–natriuresis relationship: role of the angiotensin type 2 receptor. Am J Physiol Renal Physiol 307: F901–F907. Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, Ilancheran S et al. (2009). Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Path 175: 303–313. Mookerjee I, Hewitson TD, Halls ML, Summers RJ, Mathai ML, Bathgate RA et al. (2009). Relaxin inhibits renal myofibroblast differentiation via RXFP1, the nitric oxide pathway, and Smad2. FASEB J 23: 1219–1229. Nistri S, Cinci L, Perna AM, Masini E, Bani D (2008). Mast cell inhibition and reduced ventricular arrhythmias in a swine model of acute myocardial infarction upon therapeutic administration of relaxin. Inflamm Res 57 (Suppl 1): S7–S8. Nistri S, Di Cesare ML, Ghelardini C, Zanardelli M, Bani D, Failli P (2015). Pretreatment with relaxin does not restore NO-mediated modulation of calcium signal in coronary endothelial cells isolated from spontaneously hypertensive rats. Molecules 20: 9524–9535. Ogulur I, Gurhan G, Aksoy A, Duruksu G, Inci C, Filinte D et al. (2014). Suppressive effect of compact bone-derived mesenchymal stem cells on chronic airway remodeling in murine model of asthma. Int Immunopharmacol 20: 101–109. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N et al. (2003). Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 100: 8407–8411. Parikh A, Patel D, McTiernan CF, Xiang W, Haney J, Yang L et al. (2013). Relaxin suppresses atrial fibrillation by reversing fibrosis and myocyte hypertrophy and increasing conduction velocity and sodium current in spontaneously hypertensive rat hearts. Circ Res 113: 313–321. Pearson SJ, Burgess KE, Onambélé GL (2011). Serum relaxin levels affect the in vivo properties of some but not all tendons in normally menstruating young women. Exp Physiol 96: 681–688. Perna AM, Masini E, Nistri S, Briganti V, Chiappini L, Stefano P et al. (2005). Novel drug development opportunity for relaxin in acute myocardial infarction: evidences from a swine model. FASEB J 19: 1525–1527. Petrov G, Dworatzek E, Schulze TM, Dandel M, Kararigas G, Mahmoodzadeh S et al. (2014). Maladaptive remodeling is associated with impaired survival in women but not in men after aortic valve replacement. JACC Cardiovasc Imaging 7: 1073–1080. Petrov G, Regitz-Zagrosek V, Lehmkuhl E, Krabatsch T, Dunkel A, Dandel M et al. (2010). Regression of myocardial hypertrophy after aortic valve replacement: faster in women? Circulation 122: S23–S28. Pini A, Shemesh R, Samuel CS, Bathgate RA, Zauberman A, Hermesh C et al. (2010). Prevention of bleomycin-induced pulmonary fibrosis by a novel antifibrotic peptide with relaxin-like activity. J Pharm Exp Therap 335: 589–599. Puche JE, Saiman Y, Friedman SL (2013). Hepatic stellate cells and liver fibrosis. Comprehensive Physiol 3: 1473–1492. Rivelis AR, Tregear C, Rogoff B (1965). The use of relaxin in progressive systemic sclerosis and other connective tissue diseases: a clinical study. Arch Interamerican Rheum 28: 19–31. Moore XL, Tan SL, Lo CY, Fang L, Su YD, Gao XM et al. (2007). Relaxin antagonizes hypertrophy and apoptosis in neonatal rat cardiomyocytes. Endocrinology 148: 1582–1589. Royce SG, Lim CX, Patel KP, Wang B, Samuel CS, Tang ML (2014a). Intranasally administered serelaxin abrogates airway remodelling and attenuates airway hyperresponsiveness in allergic airways disease. Clin Exp Allergy 44: 1399–1408. Mu X, Urso ML, Murray K, Fu F, Li Y (2010). Relaxin regulates MMP expression and promotes satellite cell mobilization during muscle healing in both young and aged mice. Am J Path 177: 2399–2410. Royce SG, Miao YR, Lee M, Samuel CS, Tregear GW, Tang ML (2009). Relaxin reverses airway remodeling and airway dysfunction in allergic airways disease. Endocrinology 150: 2692–2699. Nair VB, Bathgate RA, Separovic F, Samuel CS, Hossain MA, Wade JD (2015). Synthetic covalently linked dimeric form of H2 relaxin retains native RXFP1 activity and has improved in vitro serum stability. BioMed Res Int 2015: 731852. Royce SG, Moodley Y, Samuel CS (2014b). Novel therapeutic strategies for lung disorders associated with airway remodelling and fibrosis. Pharm Therap 141: 250–260. Nistri S, Chiappini L, Sassoli C, Bani D (2003). Relaxin inhibits lipopolysaccharide-induced adhesion of neutrophils to coronary endothelial cells by a nitric oxide-mediated mechanism. FASEB J 17: 2109–2111. 974 British Journal of Pharmacology (2017) 174 962–976 Royce SG, Sedjahtera A, Samuel CS, Tang ML (2013). Combination therapy with relaxin and methylprednisolone augments the effects of either treatment alone in inhibiting subepithelial fibrosis in an experimental model of allergic airways disease. Clin Sci (Lond) 124: 41–51. Anti-fibrotic actions of relaxin Royce SG, Shen M, Patel KP, Huuskes BM, Ricardo SD, Samuel CS (2015). Mesenchymal stem cells and serelaxin synergistically abrogate established airway fibrosis in an experimental model of chronic allergic airways disease. Stem Cell Res 15: 495–505. Sampson AK, Moritz KM, Jones ES, Flower RL, Widdop RE, Denton KM (2008). Enhanced angiotensin ii type 2 receptor mechanisms mediate decreases in arterial pressure attributable to chronic low-dose angiotensin ii in female rats. Hypertension 52: 666–671. Samuel CS, Bodaragama H, Chew JY, Widdop RE, Royce SG, Hewitson TD (2014). Serelaxin is a more efficacious antifibrotic than enalapril in an experimental model of heart disease. Hypertension 64: 315–322. Samuel CS, Cendrawan S, Gao XM, Ming Z, Zhao C, Kiriazis H et al. (2011). Relaxin remodels fibrotic healing following myocardial infarction. Lab Invest 91: 675–690. Samuel CS, Hewitson TD, Zhang Y, Kelly DJ (2008). Relaxin ameliorates fibrosis in experimental diabetic cardiomyopathy. Endocrinology 149: 3286–3293. Samuel CS, Lin F, Hossain MA, Zhao C, Ferraro T, Bathgate RA et al. (2007). Improved chemical synthesis and demonstration of the relaxin receptor binding affinity and biological activity of mouse relaxin. Biochemistry 46: 5374–5381. Samuel CS, Sakai LY, Amento EP (2003b). Relaxin regulates fibrillin-2 but not fibrillin-1 mRNA and protein expression in human dermal fibroblasts and fetal murine skin. Arch Biochem Biophys 411: 47–55. Samuel CS, Unemori EN, Mookerjee I, Bathgate RA, Layfield SL, Mak J et al. (2004a). Relaxin modulates cardiac fibroblast proliferation, differentiation and collagen production and reverses cardiac fibrosis in vivo. Endocrinology 145: 4125–4133. Samuel CS, Zhao C, Bathgate RA, Bond CP, Burton MD, Parry LJ et al. (2003a). Relaxin deficiency in mice is associated with an age-related progression of pulmonary fibrosis. FASEB J 17: 121–123. Samuel CS, Zhao C, Bond CP, Hewitson TD, Amento EP, Summers RJ (2004b). Relaxin-1-deficient mice develop an age-related progression of renal fibrosis. Kidney Int 65: 2054–2064. Sarwar M, Samuel CS, Bathgate RA, Stewart DR, Summers RJ (2015a). Enhanced serelaxin signalling in co-cultures of human primary endothelial and smooth muscle cells. Br J Pharmacol epub ahead of print: 23/10/2015. Sarwar M, Samuel CS, Bathgate RA, Stewart DR, Summers RJ (2015b). Serelaxin-mediated signal transduction in human vascular cells: bellshaped concentration–response curves reflect differential coupling to G proteins. Br J Pharmacol 172: 1005–1019. Sasser JM, Molnar M, Baylis C (2011). Relaxin ameliorates hypertension and increases nitric oxide metabolite excretion in angiotensin II but not N(omega)-nitro-L-arginine methyl ester hypertensive rats. Hypertension 58: 197–204. Sassoli C, Chellini F, Pini A, Tani A, Nistri S, Nosi D et al. (2013). Relaxin prevents cardiac fibroblast-myofibroblast transition via notch-1-mediated inhibition of TGF-beta/Smad3 signalling. PLoS One 8 .e63896 Scott DJ, Layfield S, Yan Y, Sudo S, Hsueh AJW, Tregear GWet al. (2006). Characterization of novel splice variants of LGR7 and LGR8 reveals that receptor signalling is mediated by their unique low density lipoprotein class A modules. J Biol Chem 281: 34942–34954. Seeger H, Lüdtke R, Gräser T, Wallwiener D, Mueck AO (2000). Effect of oral contraceptives on the urinary excretion of biochemical markers indicating vasoactive action. J Clin Pharm Ther 25: 221–226. BJP Segal MS, Sautina L, Li S, Diao Y, Agoulnik AI, Kielczewski J et al. (2012). Relaxin increases human endothelial progenitor cell NO and migration and vasculogenesis in mice. Blood 119: 629–636. Silbiger S, Neugarten J (2008). Gender and human chronic renal disease. Gend Med 5 (Suppl A): S3–S10. Seibold JR, Korn JH, Simms R, Clements PJ, Moreland LW, Mayes MD et al. (2000). Recombinant human relaxin in the treatment of scleroderma. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 132: 871–879. Singh S, Bennett RG (2010). Relaxin signalling activates peroxisome proliferator-activated receptor gamma. Mol Cell Endocrinol 315: 239–245. Singh S, Simpson RL, Bennett RG (2015). Relaxin activates peroxisome proliferator-activated receptor gamma (PPARgamma) through a pathway involving PPARgamma coactivator 1alpha (PGC1alpha). J Biol Chem 290: 950–959. Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SP et al. (2016). The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucl. Acids Res. 44: D1054–D1068. Spinale FG, Villarreal F (2014). Targeting matrix metalloproteinases in heart disease: lessons from endogenous inhibitors. Biochem Pharmacol 90: 7–15. Squecco R, Sassoli C, Garella R, Chellini F, Idrizaj E, Nistri S et al. (2015). Inhibitory effects of relaxin on cardiac fibroblast-tomyofibroblast transition: an electrophysiological study. Exp Physiol 100: 652–666. Stewart DR (2009). Scar prevention and cosmetically enhanced wound healing using relaxin. Ann N Y Acad Sci 1160: 336–341. Su W, Wang P, Chen H, Li H (2014). Role of protein kinase C beta(2) in relaxin-mediated inhibition of cardiac fibrosis. J Endocrinol Invest 37: 559–564. Teerlink JR, Metra M, Felker GM, Ponikowski P, Voors AA, Weatherley BD et al. (2009). Relaxin for the treatment of patients with acute heart failure (Pre-RELAX-AHF): a multicentre, randomised, placebocontrolled, parallel group, dose-finding phase IIb study. Lancet 373: 1429–1439. Teichman SL, Unemori E, Dschietzig T, Conrad K, Voors AA, Teerlink JR et al. (2008). Relaxin, a pleiotropic vasodilator for the treatment of heart failure. Heart Fail Rev 14: 321–329. Thien F (2013). Measuring and imaging small airways dysfunction in asthma. Asia Pacific Allergy 3: 224–230. Trautwein C, Friedman SL, Schuppan D, Pinzani M (2015). Hepatic fibrosis: concept to treatment. J Hepatol 62: S15–S24. Unemori EN, Amento EP (1990). Relaxin modulates synthesis and secretion of procollagenase and collagen by human dermal fibroblasts. J Biol Chem 265: 10681–10685. Unemori EN, Bauer EA, Amento EP (1992). Relaxin alone and in combination with interferon-gamma decreases collagen synthesis by cultured human scleroderma fibroblasts. Invest Dermatol 99: 337–3342. Unemori EN, Lewis M, Constant J, Arnold G, Grove BH, Normand J et al. (2000). Relaxin induces vascular endothelial growth factor expression and angiogenesis selectively at wound sites. Wound Repair Regen 8: 361–370. Unemori EN, Pickford LB, Salles AL, Piercy CE, Grove BH, Erikson ME et al. (1996). Relaxin induces an extracellular matrix-degrading British Journal of Pharmacology (2017) 174 962–976 975 BJP C S Samuel et al. phenotype in human lung fibroblasts in vitro and inhibits lung fibrosis in a murine model in vivo. J Clin Invest 98: 2739–2745. Vi L, Gan BS, O’Gorman DB (2010). The potential roles of cell migration and extra-cellular matrix interactions in Dupuytren’s disease progression and recurrence. Med Hypotheses 74: 510–512. Wang P, Li HW, Wang YP, Chen H, Zhang P (2009). Effects of recombinant human relaxin upon proliferation of cardiac fibroblast and synthesis of collagen under high glucose condition. J Endocrinol Invest 32: 242–247. Wang YP, Wang P, Dong L, Chen H, Wu YQ, Li HWet al. (2014). Relaxin inhibit cardiac fibrosis induced by phorbol 12-myristate 13acetate. Biomed Environmen Sci 27: 138–141. Wen Y, Zhao YY, Polan ML, Chen B (2008). Effect of relaxin on TGFbeta1 expression in cultured vaginal fibroblasts from women with stress urinary incontinence. Reprod Sci 15: 312–320. Weiss G, Teichman S, Stewart D, Nader D, Wood S, Unemori E (2009). A randomized, double-blind, placebo-controlled trial of relaxin for cervical ripening in post-delivery date pregnancies. Ann N Y Acad Sci 1160: 385–386. Wong SE, Samuel CS, Kelly DJ, Zhang Y, Becker GJ, Hewitson TD (2013). The anti-fibrotic hormone relaxin is not reno-protective, despite being active, in an experimental model of type 1 diabetes. Protein Pept Lett 20: 1029–1038. Wynn TA (2008). Cellular and molecular mechanisms of fibrosis. J Pathol 214: 199–210. Xiao J, Huang Z, Chen CZ, Agoulnik IU, Southall N, Hu X et al. (2013). Identification and optimization of small-molecule agonists of the human hormone relaxin receptor RXFP1. Nat Commun 4: 1953. Xu Q, Lekgabe ED, Gao XM, Ming Z, Tregear GW, Dart AM et al. (2008). Endogenous relaxin does not affect chronic pressure overload-induced cardiac hypertrophy and fibrosis. Endocrinology 149: 476–482. Yang JX, Zhang N, Wang HW, Gao P, Yang QP, Wen QP (2015). CXCR4 receptor overexpression in mesenchymal stem cells facilitates treatment of acute lung injury in rats. J Biol Chem 290: 1994–2006. Yoshida T, Kumagai H, Kohsaka T, Ikegaya N (2013). Relaxin protects against renal ischemia–reperfusion injury. Am J Physiol Renal Physiol 305: F1169–F1176. Williams EJ, Benyon RC, Trim N, Grove BH, Arthur MJ, Unemori EN et al. (2001). Relaxin inhibits effective collagen deposition by cultured hepatic stellate cells and decreases rat liver fibrosis in vivo. Gut 49: 577–583. Yoshida T, Kumagai H, Kohsaka T, Ikegaya N (2014). Protective effects of relaxin against cisplatin-induced nephrotoxicity in rats. Nephron Exp Nephrol 128: 9–20. Wolf JM, Cameron KL, Clifton KB, Owens BD (2013). Serum relaxin levels in young athletic men are comparable with those in women. Orthopedics 36: 128–131. Zhang X, Ma X, Zhao M, Zhang B, Chi J, Liu Wet al. (2015). H2 and H3 relaxin inhibit high glucose-induced apoptosis in neonatal rat ventricular myocytes. Biochimie 108: 59–67. 976 British Journal of Pharmacology (2017) 174 962–976