HHS Public Access Author manuscript Author Manuscript

Kidney Int. Author manuscript; available in PMC 2017 June 01. Published in final edited form as: Kidney Int. 2016 June ; 89(6): 1231–1243. doi:10.1016/j.kint.2016.02.002.

Ligand trap for the Activin Type IIA receptor protects against vascular disease and renal fibrosis in mice with chronic kidney disease Olga A. Agapova, Ph.D.1, Yifu Fang, M.D.1, Toshifumi Sugatani, D.D.S., Ph.D.1, Michael E. Seifert, M.D.1,3, and Keith A. Hruska, M.D.1,2,*

Author Manuscript

1Department

of Pediatrics, Renal Division, Washington University, St. Louis, MO

2Department

of Cell Biology, and Medicine, Washington University, St. Louis, MO

3Department

of Pediatrics, Renal Division, Southern Illinois University, Springfield, IL

Abstract

Author Manuscript

The causes of cardiovascular mortality associated with chronic kidney disease (CKD) are partly attributed to the CKD-mineral bone disorder (CKD-MBD). The causes of the early CKD-MBD are not well known. Our discovery of Wnt (portmanteau of wingless and int) inhibitors, especially Dickkopf 1, produced during renal repair as participating in the pathogenesis of the vascular and skeletal components of the CKD-MBD implied that additional pathogenic factors are critical. In the search for such factors, we studied the effects of activin receptor type II A (ActRIIA) signaling by using a ligand trap for the receptor, RAP-011 (a soluble extracellular domain of ActRIIA fused to a murine IgG-Fc fragment). In a mouse model of CKD that stimulated atherosclerotic calcification, RAP-011 significantly increased aortic ActRIIA signaling assessed by the levels of phosphorylated Smad2/3. Furthermore, RAP-011 treatment significantly reversed CKD induced vascular smooth muscle dedifferentiation as assessed by smooth muscle 22α levels, osteoblastic transition and neointimal plaque calcification. In the diseased kidneys, RAP-011 significantly stimulated αklotho levels and it inhibited ActRIIA signaling and decreased renal fibrosis and proteinuria. RAP-011 treatment significantly decreased both renal and circulating Dickkopf 1 levels showing that Wnt activation was downstream of ActRIIA. Thus, ActRIIA signaling in CKD contributes to the CKD-MBD and renal fibrosis. ActRIIA signaling may be a potential therapeutic target in CKD.

Author Manuscript

Keywords chronic kidney disease; vascular calcification; fibrosis; signaling

*

Corresponding Author Keith A. Hruska MD, Renal Division, Department of Pediatrics, Washington University, Rm 5109 MPRB Building, 660 S. Euclid, Saint Louis, MO 63110, Phone: 314-286-2772, [email protected]. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: The other authors declare no conflicts of interest exist.

Agapova et al.

Page 2

Author Manuscript

Introduction

Author Manuscript

Kidney diseases are associated with high mortality rates related to their stimulation of cardiovascular disease.1 The kidney disease stimulation of cardiovascular risk extends to type 2 diabetes, where the presence of mild to moderate kidney disease increases atherosclerotic cardiovascular disease risk by 87%.2 Atherosclerotic calcification has become a standard biomarker of cardiovascular risk,3, 4 and CKD increases atherosclerotic calcification.5 The causes of the increased cardiovascular risk associated with kidney diseases partly reside in the chronic kidney disease – mineral bone disorder (CKD-MBD) syndrome.6 Three non-traditional cardiovascular risk factors (hyperphosphatemia, vascular calcification, and elevated fibroblast growth factor 23 (FGF23) levels) have been discovered in the CKD-MBD,7-9 and their risk factor status confirmed in the general population.10-12 The CKD-MBD begins early in CKD (stage 2)13-16 consisting of arterial vascular cell dedifferentiation/calcification, an osteodystrophy, loss of klotho and increased FGF23 secretion.13 Progress into the causes of the CKD-MBD has been made,16-19 but they are mostly unknown. We have employed a murine model of type 2 diabetes, atherosclerosis and atherosclerotic calcification to demonstrate that CKD stimulates atherosclerotic calcification.20, 21 The development of hyperphosphatemia further stimulates the process,22, 23 but is not involved in its inception which begins much earlier in the course of CKD than hyperphosphatemia. Most recently we have focused on circulating factors produced by kidney disease that perturb normal physiologic systemic processes.

Author Manuscript

Multiple investigators and we have shown that kidney diseases reactivate developmental programs involved in nephrogenesis during disease stimulated renal repair.24-28 Among the nephrogenic factors reactivated in renal repair, the Wnt (portmanteau of Wingless and Integrated) family is critical for tubular epithelial reconstitution,27-29 and fibrosis. In the control of Wnt function, canonical signaling transcriptionally induces the expression of a family of Wnt inhibitory proteins which are secreted proteins that serve to restrict the distances of Wnt stimulation to autocrine or paracrine factors.30-34 The Wnt inhibitors are circulating factors, and the family includes the Dickkopfs (Dkk). We have shown that various forms of kidney disease increase renal expression of Wnt inhibitors including the Dkk family and increase their levels in the systemic circulation.17, 25

Author Manuscript

Neutralization of a key Wnt inhibitor elevated in the circulation in CKD, Dkk1, was efficacious in the CKD-MBD. Dkk1 neutralization inhibited CKD induced vascular dedifferentiation, vascular calcification, and renal osteodystrophy.17 This effect was surprising since Wnt signaling in the vascular smooth muscle cell (VSMC) is implicated in stimulating osteoblastic transition and vascular calcification.35, 36 However, recent studies demonstrate that inhibition of Wnt signaling stimulates lipid accumulation in atherosclerotic plaques,37 and that Dkk1 mediated inhibition of aortic Wnt7b stimulates smad mediated aortic endothelial-mesenchymal transition (EndMT) and vascular calcification.38 EndMT is a developmental physiologic process involved in the development of the cardiac valves, the cardiac septum and the aortic root,39, 40 and it may41 or may not42 contribute to cardiac fibrosis in various adult disease states. Since EndMT is a process driven by Smad transcription factors activated by receptors for the transforming growth factor beta (TGFβ) superfamily,43 we investigated whether changes in TGFβ superfamily receptor function are

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 3

Author Manuscript

involved in the pathogenesis of the CKD-MBD. Our initial strategy was to focus on the function of putatively important TGFβ superfamily receptors. The superfamily ligands generally bind to type two receptors which associate and activate type one receptors initiating signal transduction. Besides TGFβ receptor II (TGFβRII), bone morphogenetic protein type 2 receptor (BMPRII), activin receptor type 2A (ActRIIA) and activin receptor type 2B (ActRIIB) are the most important type 2 receptors of the superfamily.

Author Manuscript

Here we report the effects of a ligand trap for the activin type 2A receptor, ActRIIA, on CKD stimulated atherosclerotic calcification and renal αklotho levels, because the latter is a component of the CKD-MBD and has been implicated in vascular calcification.44 Aortic VSMC ActRIIA levels and ActRIIA signaling were decreased by CKD. The ActRIIA ligand trap increased aortic ActRIIA signaling measured by Smad activation, blocked CKD stimulated vascular smooth muscle osteoblastic transition, and decreased atherosclerotic vascular calcification. In the kidney, we found that the ligand trap increased renal αklotho expression. Furthermore in the kidney, ActRIIA levels were not changed by CKD, and the ligand trap decreased renal ActRIIA signaling. The ligand trap also decreased renal Wnt activation and decreased circulating Dkk1. Renal fibrosis and proteinuria were decreased by the ActRIIA ligand trap. The compiled result was that RAP-011 decreased vascular calcification, and renal fibrosis.

Results Experimental Design, and Kidney function in a model of CKD

Author Manuscript

To study the effect of the ActRIIA ligand trap on the CKD-MBD in early CKD, we used the high fat fed ldlr−/− mouse with ablative CKD treated with a ligand trap for the receptor. The experimental design of the ActRIIA ligand trap experiments in our model is shown in Figure 1. The high fat fed ldlr−/− mouse is a well characterized model of atherosclerotic vascular calcification requiring both the diet and the genotype to produce atherosclerotic vascular calcification.20, 45 We have used the model extensively to study stimulation of atherosclerotic calcification by CKD.13, 17, 20, 21, 23, 46-48 In the present studies, kidney function was reduced to approximately 30% of normal (CKD) in ldlr−/− high fat fed mice by renal cortical injury and contralateral nephrectomy (supplemental Figure 1). The CKD mice were hyperphosphatemic (Table 1) Activin Receptor Type IIA (ActRIIA) levels in CKD

Author Manuscript

Aortas from the high fat fed ldlr−/− mice with CKD were analyzed for TGFβ superfamily type II receptors which are the ligand binding component of the superfamily receptor heteromultimers composed of type II and type I (ALK) receptors. The Activin type II receptor A (ActRIIA) was expressed in aortic vascular smooth muscle cells (VSMC) of the ldlr−/− high fat fed mice (Fig. 2A,B). CKD induced ActRIIA down regulation in the aorta (Fig. 2A,B). This is consistent with internalization and degradation of ActRIIA produced by high circulating ligand levels reported in other tissues.49, 50 Endothelial cell ActRIIA was not detected by immunochemical and immunofluorescent detection. (Fig. 2B).

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 4

Vascular effects of the ActRIIA ligand trap in CKD

Author Manuscript Author Manuscript

The effects of CKD-induced suppression of ActRIIA levels were analyzed in aortic homogenates from CKD mice treated with vehicle or the ActRIIA ligand trap (RAP-011) (Fig. 3). First, CKD stimulated osteoblastic transition was assessed by expression of mRNA for Runx2 and alkaline phosphatase (Alpl) in the aortas of ldlr−/− high fat fed mice. CKD stimulated their expression and RAP-011 treatment reversed the effects of CKD (Fig. 3A). Both Runx2 and Alpl expression represent biomarkers of osteoblastic transition in the aorta that were reversed by RAP-011 treatment. Next, aortic mRNA of smooth muscle 22α or transgelin (Tagln), a biomarker of differentiated vascular smooth muscle cells, 51 was decreased by CKD and stimulated by RAP-011. CKD also caused decreased aortic myocardin (Myocd) mRNA expression, the vascular smooth muscle cell specific transcription factor, but myocardin was not affected by RAP-011 treatment. In terms of the effects of CKD and RAP-011 treatment on aortic protein levels of the respective mRNAs studied in Figure 3A, CKD increased aortic Runx2 and Alpl levels and RAP-011 normalized them (Fig. 3B, data for Alpl not shown). CKD decreased the aortic levels of Tagln and alpha smooth muscle actin (αSMA), another biomarker of differentiated vascular smooth muscle cells, and RAP-011 treatment increased them (Fig. 3B, data for Tagln not shown). Myocardin levels were not changed by CKD or RAP-011 treatment.

Author Manuscript

The ldlr−/− high fat fed mouse has atherosclerotic calcification35, 45 stimulated by CKD as described above. In the studies reported here, CKD caused accumulation of calcium deposits in the aortic atheromas in CKD vehicle treated mice (CKD V) (Fig. 4A) and increased total tissue calcium levels (Fig. 4B). Visible calcium deposits were not present in CKD mice treated with RAP-011 (CKD RAP-011), and RAP-011 decreased aortic tissue calcium content to levels observed in wild type and sham mice, significantly below those present at the time of institution of RAP-011 treatment, the CKD group (Fig. 4B). The dose of RAP-011 used here, 10mg/kg twice weekly subcutaneously, was used to relate to prior toxicology, pharmocodynamic and efficacy studies. Subsequent to the studies in Figure 4B, we performed a dose response study and equivalent or better reductions in aortic tissue calcium content resulted when doses of 5mg/kg and 1mg/kg twice weekly subcutaneously were used (data not shown). ActRIIA signaling in the aorta

Author Manuscript

Despite the decrease in ActRIIA in CKD, there remained easily detected ActRIIA in the VSMC potentially available for signaling (Fig. 2B). Canonical signal transduction by the TGFβ superfamily involves ligand binding to type II receptors activating their serine/ threonine kinase activity and stimulating association and phosphorylation of type I receptors, the Alk kinases (see diagrammatic representation in supplemental Figure 2). There are seven Alk kinases utilized by the TGFβ superfamily, and Alk4 (ActRIB) is the type I receptor most often associated with ActRIIA signaling.52 Aortic homogenates isolated from CKD mice revealed that the decrease in ActRIIA levels, which were shown quantatively in Figure 2 above, were not associated with decreased tissue levels of Alk4, and Alk1 (Fig. 5A). Alk5 or Alk2, other type 1 receptors associated with ActRIIA signaling, were not detectable. ActRIIA activity was assessed by measuring the effect of receptor heteromultimerization, i.e., phosphorylation of regulatory Smads. CKD decreased aortic Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 5

Author Manuscript Author Manuscript

phosphosmad 2/3 levels (activated smad 2/3), and RAP-011 increased them compared to CKDV, even though total Smad2/3 levels were decreased (Fig. 5A, B). We also analyzed noncanonical ActRIIA signaling (supplemental Figure 2), and found that map kinase (phospho-Erk1/2) was also decreased by CKD and not further affected by RAP-011 (Fig. 5A), and that vascular smooth muscle levels of p38 and JNK were very low. Also, RAP-011 did not affect p-AKT levels indicating that aortic AKT/ PI3 kinase was not affected by ActRIIA signaling (data not shown). In summary, aortic ActRIIA signaling (phosphosmad 2/3) was decreased by CKD and stimulated by the ligand trap associated with suppression of osteoblastic transition in the atherosclerotic aortas by the ligand trap (Fig. 3). When RAP-011 was administered to wild type mice, the reduction in total Smad 2/3 levels observed in CKD mice were reproduced (Fig 5C). However, WT mice had very low levels of ActRIIA and p-Smad 2/3 which were not affected by RAP-011. RAP-011 was well tolerated in mice with normal kidney function,53 and there was no discernible effect of RAP-011 on appearance, activity, food intake, and weight gain on the CKD mice. There was the expected increase in hematocrit, but this was an efficacious side effect.

Author Manuscript

Because the effects of RAP-011 treatment on the aorta were similar to those of Dkk1 inhibition,17 another non-canonical ActRIIA signaling pathway was examined, i.e. - the Wnt pathway (supplemental Figure 2). Aortic β-catenin, the major canonical Wnt induced transcription factor, was localized by immunofluorescence to endothelial cells, and was not detectable in VSMC (Fig. 6A). But because Dkk1 is a transcriptional target of canonical Wnt signaling, and its mRNA is detectable in VSMC in vivo reflecting low level Wnt activity, we analyzed Dkk1 levels in the VSMC as a biomarker of Wnt activity. We found that CKD increased VSMC Dkk1 levels, and RAP-011 treatment decreased Dkk1 levels in our CKD mice (Fig. 6B). The decrease in aortic Dkk1 levels by RAP-011 suggests that vascular smooth muscle Wnt signaling was inhibited by the ActRIIA ligand trap. In addition, the effects of the ligand trap to decrease renal Wnt activity as shown below lead to a major decrease in circulating Dkk1 levels (Fig. 6C). 54 Effects of the ActRIIA ligand trap on αklotho and renal ActRIIA signaling

Author Manuscript

Because αklotho is a critical component of the CKD-MBD and a pathogenic factor in vascular calcification,16, 55, 56 we examined renal αklotho levels and the effects of CKD and RAP-011 treatment. Recent studies demonstrate that kidney tubules are the source of most αklotho.57 As shown in Figure 7A, we found a significant increase in renal αklotho levels induced by RAP-011 treatment. Therefore, we studied renal ActRIIA levels and ActRIIA signaling to examine whether the effect of CKD to downregulate the receptor and decrease signaling observed in the aorta were reproduced in the kidney. Renal ActRIIA levels were not affected by CKD (Fig.7B), and a primary ActRIIA ligand, activin A, was strongly induced in the CKD mice, and was suppressed by RAP-011 treatment (quantitation of activin A levels is shown below in Fig. 9). Detecting type I receptor activation was impaired by the lack of available phospho-Alk antibodies, requiring an experimental design wherein a phosphoserine antibody was used to immunoprecipitate and immunoblots of the precipitate were performed with anti-Alk antibodies. We were unable to do this in the aorta due to insufficient amounts of tissue, but we were able to perform these experiments using kidney homogenates. Alk4 phosphorylation was not significantly altered in the diseased kidneys of

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 6

Author Manuscript

our CKD model or by RAP-011 treatment (Fig. 7B), but renal phosphosmad 2/3 was increased by CKD and decreased by RAP-011 (Fig. 7B,C) indicating that the component of renal Smad 2/3 activation in CKD mediated through ActRIIA involved an Alk different from Alk4 (Fig. 7B). Furthermore, psmad 2/3 transcriptional targets, fibronectin and type 1 collagen (Col1A1) were stimulated by CKD and inhibited by RAP-011 treatment (Fig. 7B,C) .

Author Manuscript

Because increased pSmad 3 is associated with renal fibrosis, and since ActRIIA signaling stimulated by activin A is a critical factor in fibrosis 58, 59 including renal fibrosis, 60, 61 we examined the effects of the ActRIIA ligand trap on renal fibrosis. Figure 8A shows the decrease in renal fibrosis in Trichrome stained kidney cortex sections from CKD RAP-011 treated mice (c and d) compared to CKDV treated mice (a and b). Supplemental Figure 3 shows low magnification coronal remnant kidney sections with arrows showing from where the high power sections in Figure 8A were taken, away from the scar reactions to the electrocautery delineate by arrow heads. Quantitation of fibrosis by interstitial volume analysis is shown in Figure 8B. Furthermore, RAP-011 decreased the proteinuria stimulated by CKD (Fig.8C) consistent with the decrease in renal phosphosmad 2/3 and fibrosis induced by RAP-011 treatment. Potential ActRIIA ligands in CKD

Author Manuscript Author Manuscript

There are multiple potential ActRIIA ligands including activins A and B, growth and differentiation factor 11 (GDF11), bone morphogenetic proteins 9 and 10 (BMP9 and 10), and other BMPs, especially 7, which have lower affinity for the receptor. But a primary ActRIIA ligand is activin. We do not know the ligand/s involved in disordered ActRIIA signaling in CKD, which we have just begun to study as a result of the findings reported here. Using two models of CKD, we found that systemic circulating activin A levels were 10 fold elevated in the high fat fed ldlr−/− ablative CKD model and fivefold elevated in the Col4A5 Alport's syndrome mouse model (Fig. 9A). Activin B, GDF11, and BMP9 levels were not affected by CKD in these models. Physiologically, there is little free activin in the circulation due to levels of inhibitors stoichiometrically equaling activin levels. Activins associate with circulating inhibitory factors, follistatin and follistatin like 3 (fstL3),62 and inhibin whose circulating levels (supplemental Figure 4) and tissue levels were not affected or decreased by CKD. The stoichiometry of follistatin, fstL3, and inhibin (sum of 620pg/ml in supplemental figure 4, plus 400pg/ml of unmeasured inhibin 63) to Activin A levels (>5000pg/ml) in the circulation suggests that CKD produces significant free activin levels, a pathologic event making activin A an active circulating factor in CKD. While the other potential ActRIIA ligands affected by CKD require additional study, this data indicates the potential that kidney diseases produce one or more circulating ActRIIA ligands that could potentially downregulate vascular ActRIIA. To analyze the source of the increase in circulating activin, we analyzed kidney tissues from our ldlr−/− atherosclerotic calcification model for activin A (a homodimer of Inhibin betaA (Inhba)). Inhba mRNA was increased by CKD in the kidney (Fig. 9B), and activin (inhibin β-A) protein levels were increased (Fig. 9C). Localization of renal activin expression was found in the peritubular myofibroblasts of CKD mice (Fig. 10). However, these data do not

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 7

Author Manuscript

establish that activin is the ligand involved in disordered ActRIIA signaling in CKD, and additional research involving production of myofibroblast activin deficient mice and ActRIIA cell specific deficient mice will be necessary to establish this issue.

Discussion

Author Manuscript Author Manuscript

Our results report reversal of CKD induced stimulation of aortic vascular smooth muscle dedifferentiation, osteoblastic transition and neointimal calcification by a ligand trap for ActRIIA, RAP-011. These results build on our recent demonstration that kidney diseases increase circulating Wnt inhibitors, that systemic Wnt inhibition contributes to CKD induced vascular and skeletal disease, and that neutralization of elevated Dkk1 prevents the vascular calcification and renal osteodystrophy of early CKD.17 The effects of Dkk1 neutralization to decrease vascular calcification were surprising because several investigators had linked vascular smooth muscle Wnt activity to vascular calcification.35, 36, 64 However, the recent demonstration that Dkk1 mediated inhibition of Wnt7b caused EndMT,38 lead us to question whether EndMT was a mechanism of vascular disease and vascular calcification in CKD. EndMT is a Smad mediated process. 43 Regulatory Smads are transcription factors activated by members of the TGFβ superfamily that includes the TGFβs, bone morphogenetic proteins (BMPs), activins/inhibins, and growth and differentiation factors (GDFs). 65 We employed a ligand trap for the ActRIIA receptor to interrogate the role of Smad stimulated pathways in the vasculature during CKD. We found that treatment with the ActRIIA ligand trap decreased atherosclerotic calcification in the aorta, decreased expression of the osteoblastic proteins, Runx2 and Alpl, and increased levels of smooth muscle cell specific proteins, sm22α and αSMA (alpha smooth muscle actin). Since the RAP-011 ligand trap increased aortic p-Smad 2/3 levels, these data suggest that downregulation of vascular ActRIIA signaling in CKD contributes to altered VSMC biology involved in upregulation of Runx2 and osteochondrogenic transition critical to atherosclerotic calcification.66, 67 When we attempted to study this biology in VSMC cell culture we discovered that murine VSMCs in culture do not express ActRIIA protein despite the mRNA being transcribed (data not shown). This again is consistent with the dedifferentiated state that characterizes VSMC cell cultures and suggests that ActRIIA signaling may be important in VSMC differentiation as our data in vivo suggests (the ligand trap treatment increased VSMC sm22α (tagln) and αSMA).

Author Manuscript

We found that CKD was associated with reduction of ActRIIA protein levels in the aortas of our mouse model of atherosclerotic calcification, and that this correlated with decreased activated Smad 2/3 levels (phosphosmad 2/3) and decreased MAP kinase (phosphoERK 1/2) indicating that CKD produced inhibition of aortic ActRIIA signaling. The finding that the ligand trap actually increased ActRIIA signaling (increased aortic p-Smad 2/3 levels) suggests that ActRIIA was suppressed in CKD mice due to high levels of a ligand inducing receptor desensitization possibly through endocytosis and degradation (supplemental Figure 5). The ActRIIA receptor undergoes ligand stimulated endocytosis during signal transduction, 68 and can be down regulated during the signal transduction process depending on the effects of activin receptor interacting proteins (ARIP1 and ARIP2). 68 While ARIP1 and ARIP2b and 2c have been associated with Smad binding and effective signaling,69

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 8

Author Manuscript

ARIP2 is associated with receptor endocytosis, stimulation of receptor degradation and decreased signaling.50, 68 Downregulation of vascular ActRIIA by ligand stimulated endocytosis is consistent with the decrease in aortic ActRIIA in the CKD mice because of the high levels of circulating activin A produced by CKD. High activin levels have been shown to stimulate endocytotic mediated degradation of ActRIIA through the effects of ARIP2.68 In addition, ARIP2 is more highly expressed in the vasculature related to ARIP1, while in the kidney ARIP1 expression is much higher than ARIP2 (supplemental Figure 6). Results similar to the VSMC effects of CKD on ActRIIA levels, have been produced in testicular cells exposed to high activin levels.49 These data suggest that CKD may produce vascular ActRIIA downregulation through production of a ligand in the circulation since vascular tissue ligands of ActRIIA were also downregulated in CKD as shown for the primary ActRIIA ligand, activin A (Fig. 5). This led us to analyze ActRIIA signaling and potential ligands in the diseased kidney.

Author Manuscript Author Manuscript

In the kidney we found that ActRIIA levels were not affected by CKD or RAP-011 treatment consistent with a different ARIP involved in renal ActRIIA signaling than that involved in the aorta consistent with the high level of renal ARIP1 mRNA shown in supplemental Figure 6. In the kidney, there was a major induction of activin A, a primary ligand for ActRIIA, by CKD and a coordinate increase in activated Smad 2/3 (p-Smad 2/3). RAP-011 treatment decreased renal activin A expression and decreased p-Smad 2/3. This indicates that in the kidney ActRIIA signaling was increased by CKD and reduced by RAP-011 treatment. Furthermore, CKD induced Wnt activation measured by increased renal levels of Dkk1 was inhibited by RAP-011. In agreement with these results, renal fibrosis, proteinuria, type 1 collagen levels and fibronectin levels were increased by CKD consistent with the effects of increased p-Smad 3 induced gene transcription, and decreased by RAP-011 treatment. Importantly, RAP-011 treatment increased renal production of αklotho, which may have contributed to its efficacy in inhibiting atherosclerotic vascular calcification.16

Author Manuscript

In the diseased kidney, the profibrogenic effects of Wnt activation are marked by increased expression of the Wnt inhibitor, Dkk1.24, 25, 28 In the diseased kidney, RAP-011 treatment decreased kidney expression of Dkk1 and decreased circulating Dkk1 levels. These effects are consistent with cooperative effects of activin and the Wnt pathways during development 70-73 and kidney disease as shown here. Thus, we have shown that reactivation of two kidney development factors, activin and Wnt in kidney disease produce circulating Wnt inhibitors and activin in CKD, which may interact in causing vascular disease. Our data are consistent but do not prove that renal production of a circulating ActRIIA ligand was the basis for the downregulation of vascular ActRIIA signaling which was stimulated by RAP-011 treatment, and they do not identify the ActRIIA ligand affected by RAP-011 further than indicating that activin A is a candidate for this function. However, the ActRIIA ligand trap, decreased Dkk1 levels linking the findings reported here to our previous report on Dkk1 inhibition. In conclusion, we report the discovery of disordered ActRIIA signaling during CKD induced atherosclerotic calcification and renal fibrosis. The results demonstrate that a ligand trap for the receptor improved ActRIIA signaling, and smooth muscle specific protein expression in Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 9

Author Manuscript

the aorta. It decreased osteoblastic transition, and decreased atherosclerotic vascular calcification. RAP-011 also increased renal αklotho expression and decreased renal fibrosis. Secondarily, RAP-011 decreased renal Wnt signaling and circulating Dkk1, which contributed to the efficacy of RAP-011 in restoring vascular function during CKD. Since the human ortholog of RAP-011 (sotatercept) is in clinical trials for the anemia of CKD (NCT01146574, NCT01999582), and ACE-011 has been shown to be effective in reducing elevated GDF-11 levels in β-thalassemia 74, there may be opportunity for further exploring the efficacy of ActRIIA ligand traps in the CKD-MBD. Thus, we have shown that regulation of ActRIIA signaling in CKD may be a new therapeutic target modulating vascular disease and kidney disease progression.

Methods Author Manuscript

The studies reported here used methodology previously reported and described in detail in the supplementary materials.

Supplementary Material Refer to Web version on PubMed Central for supplementary material.

Acknowledgments Thanks to Helen Odle who assisted in preparation of the manuscript. These studies were supported by NIH grants DK070790 (KAH) and DK089137 (KAH), and P30 AR057235 core center for musculoskeletal biology and medicine. Additional support was in the form of an investigator stimulated trial grant from Celgene (KAH)

Author Manuscript

KAH declares that the work presented in the manuscript was supported by NIH grants DK070790 and DK089137, and by investigator stimulated trial grants from Celgene. Celgene is developing Sotatercept as a therapy for the anemia of CKD. In addition, he is an advisor for Celgene. Sources of Support: NIH grants DK070790 and DK089137, and investigator stimulated trial grants from Celgene

References

Author Manuscript

1. Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney Disease as a Risk Factor for Development of Cardiovascular Disease. Circulation. 2003; 108:2154–2169. [PubMed: 14581387] 2. Papademetriou V, Lovato L, Doumas M, et al. Chronic kidney disease and intensive glycemic control increase cardiovascular risk in patients with type 2 diabetes. Kidney Int. 2015; 87:649–659. [PubMed: 25229335] 3. Pletcher MJ, Tice JA, Pignone M, et al. Using the Coronary Artery Calcium Score to Predict Coronary Heart Disease Events: A Systematic Review and Meta-analysis. Arch Intern Med. 2004; 164:1285–1292. [PubMed: 15226161] 4. Wexler L, Brundage B, Crouse J, et al. Coronary artery calcification: Pathophysiology, epidemiology, imaging methods, and clinical implications. Circulation. 1996; 94:1175–1192. [PubMed: 8790070] 5. Barraclough KA, Stevens LA, Er L, et al. Coronary artery calcification scores in patients with chronic kidney disease prior to dialysis: reliability as a trial outcome measure. Nephrol Dial Transplant. 2008; 23:3199–3205. [PubMed: 18450828] 6. Moe S, Drueke T, Cunningham J, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from kidney disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006; 69:1945–1953. [PubMed: 16641930]

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 10

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

7. Block GA, Hulbert-Shearon TE, Levin NW, et al. Association of serum phosphorus and calcium X phosphate product with mortality risk in chronic hemodialysis patients: a national study. Am J Kidney Dis. 1998; 31:607–617. [PubMed: 9531176] 8. Blacher J, Guerin AP, Pannier B, et al. Arterial calcifications, arterial stiffness, and cardiovascular risk in end-stage renal disease. Hypertension. 2001; 38:938–942. [PubMed: 11641313] 9. Gutierrez OM, Mannstadt M, Isakova T, et al. Fibroblast Growth Factor 23 and Mortality among Patients Undergoing Hemodialysis. New Engl J Med. 2008; 359:584–592. [PubMed: 18687639] 10. Dhingra R, Sullivan LM, Fox CS, et al. Relations of Serum Phosphorus and Calcium Levels to the Incidence of Cardiovascular Disease in the Community. Arch Intern Med. 2007; 167:879–885. [PubMed: 17502528] 11. Matsushita K, Sang Y, Ballew SH, et al. Subclinical Atherosclerosis Measures for Cardiovascular Prediction in CKD. J Am Soc Nephrol. 2014; 26:439–447. [PubMed: 25145930] 12. Dalal M, Sun K, Cappola AR, et al. Relationship of serum fibroblast growth factor 23 with cardiovascular disease in older community-dwelling women. Eur J Endocrinol. 2011; 165:797– 803. [PubMed: 21873490] 13. Fang Y, Ginsberg C, Sugatani T, et al. Early chronic kidney disease-mineral bone disorder stimulates vascular calcification. Kidney Int. 2014; 85:142–150. [PubMed: 23884339] 14. Pereira RC, Juppner H, Azucena-Serrano CE, et al. Patterns of FGF-23, DMP1 and MEPE expression in patients with chronic kidney disease. Bone. 2009; 45:1161–1168. [PubMed: 19679205] 15. Fang Y, Zhang Y, Mathew S, et al. Early chronic kidney disease (CKD) stimulates vascular calcification (VC) and decreased bone formation rates prior to positive phosphate balance. J Am Soc Nephrol. 2009; 20:36A. 16. Hu MC, Shi M, Zhang J, et al. Klotho Deficiency Causes Vascular Calcification in Chronic Kidney Disease. J Am Soc Nephrol. 2011; 22:124–136. [PubMed: 21115613] 17. Fang Y, Ginsberg C, Seifert M, et al. CKD-induced Wingless/Integration1 Inhibitors and Phosphorus Cause the CKD-Mineral and Bone Disorder. J Am Soc Nephrol. 2014; 25:1760–1763. [PubMed: 24578135] 18. de Oliveira RB, Graciolli FG, dos Reis LM, et al. Disturbances of Wnt/β-catenin pathway and energy metabolism in early CKD: effect of phosphate binders. Nephrol Dial Transplant. 2013; 28:2510–2517. [PubMed: 23975746] 19. Sabbagh Y. Repression of osteocyte Wnt/[beta]-catenin signaling is an early event in the progression of renal osteodystrophy. J Bone Miner Res. 2012; 27:1757–1772. [PubMed: 22492547] 20. Davies MR, Lund RJ, Hruska KA. BMP-7 is an efficacious treatment of vascular calcification in a murine model of atherosclerosis and chronic renal failure. J Am Soc Nephrol. 2003; 14:1559– 1567. [PubMed: 12761256] 21. Mathew S, Lund R, Strebeck F, et al. Reversal of the adynamic bone disorder and decreased vascular calcification in chronic kidney disease by sevelamer carbonate therapy. J Am Soc Nephrol. 2007; 18:122–130. [PubMed: 17182886] 22. El-Abbadi MM, Pai AS, Leaf EM, et al. Phosphate feeding induces arterial medial calcification in uremic mice: role of serum phosphorus, fibroblast growth factor-23, and osteopontin. Kidney Int. 2009; 75:1297–1307. [PubMed: 19322138] 23. Mathew S, Tustison KS, Sugatani T, et al. The mechanism of phosphorus as a cardiovascular risk factor in chronic kidney disease. J Am Soc Nephrol. 2008; 19:1092–1105. PMCID:PMC2396927. [PubMed: 18417722] 24. Surendran K, McCaul SP, Simon TC. A role for Wnt-4 in renal fibrosis. Am J Physiol Renal Physiol. 2002; 282:F431–F441. [PubMed: 11832423] 25. Surendran K, Schiavi S, Hruska KA. Wnt-dependent-β-catenin signaling is activated after unilateral ureteral obstruction, and recombinant secreted frizzled-related protein 4 alters the progression of renal fibrosis. J Am Soc Nephrol. 2005; 16:2373–2384. [PubMed: 15944336] 26. Maeshima A, Nojima Y, Kojima I. The role of the activin-follistatin system in the developmental and regeneration processes of the kidney. Cytokine & Growth Factor Reviews. 2001; 12:289–298. [PubMed: 11544099]

Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 11

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

27. Terada Y, Tanaka H, Okado T, et al. Expression and Function of the Developmental Gene Wnt-4 during Experimental Acute Renal Failure in Rats. J Am Soc Nephrol. 2003; 14:1223–1233. [PubMed: 12707392] 28. Kawakami T, Ren S, Duffield JS. Wnt signalling in kidney diseases: dual roles in renal injury and repair. J Pathol. 2013; 229:221–231. [PubMed: 23097132] 29. Rinkevich Y, Montoro Daniel T, Contreras-Trujillo H, et al. In Vivo Clonal Analysis Reveals Lineage-Restricted Progenitor Characteristics in Mammalian Kidney Development, Maintenance, and Regeneration. Cell Reports. 2014; 7:1270–1283. [PubMed: 24835991] 30. Niida A, Hiroko T, Kasai M, et al. DKK1, a negative regulator of Wnt signaling, is a target of the á-catenin/TCF pathway. Oncogene. 2004; 23:8520–8526. [PubMed: 15378020] 31. Niehrs C. Function and biological roles of the Dickkopf family of Wnt modulators. Oncogene. 2006; 25:7469–7481. [PubMed: 17143291] 32. Reya T, Duncan AW, Ailles L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003; 423:409–414. [PubMed: 12717450] 33. Chamorro MN, Schwartz DR, Vonica A, et al. FGF-20 and DKK1 are transcriptional targets of βcatenin and FGF-20 is implicated in cancer and development. EMBO J. 2004; 24:73–84. [PubMed: 15592430] 34. Gonzalez-Sancho JM, Aguilera O, Garcia JM, et al. The Wnt antagonist DICKKOPF-1 gene is a downstream target of [beta]-catenin//TCF and is downregulated in human colon cancer. Oncogene. 2004; 24:1098–1103. [PubMed: 15592505] 35. Al-Aly Z, Shao JS, Lai CF, et al. Aortic Msx2-Wnt Calcification Cascade Is Regulated by TNF{alpha} Dependent Signals in Diabetic Ldlr / Mice. Arterioscler Thromb Vasc Biol. 2007; 27:2589–2596. [PubMed: 17932314] 36. Shao JS, Cheng SL, Pingsterhaus JM, et al. Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J Clin Invest. 2005; 115:1210–1220. [PubMed: 15841209] 37. Borrell-Pagès M, Romero JC, Badimon L. LRP5 deficiency down-regulates Wnt signalling and promotes aortic lipid infiltration in hypercholesterolaemic mice. J Cell Mol Med. 2015; 19:770– 777. [PubMed: 25656427] 38. Cheng S-L, Shao J-S, Behrmann A, et al. Dkk1 and Msx2–Wnt7b Signaling Reciprocally Regulate the Endothelial–Mesenchymal Transition in Aortic Endothelial Cells. Arterio Thromb Vasc Biol. 2013; 33:1679–1689. 39. Eisenberg LM, Markwald RR. Molecular Regulation of Atrioventricular Valvuloseptal Morphogenesis. Circ Res. 1995; 77:1–6. [PubMed: 7788867] 40. Camenisch TD, Molin DGM, Person A, et al. Temporal and Distinct TGFβ Ligand Requirements during Mouse and Avian Endocardial Cushion Morphogenesis. Develop Biol. 2002; 248:170–181. [PubMed: 12142029] 41. Zeisberg EM, Tarnavski O, Zeisberg M, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007; 13:952–961. [PubMed: 17660828] 42. Moore-Morris T, Guimar xE, et al. Resident fibroblast lineages mediate pressure overload–induced cardiac fibrosis. J Clin Invest. 2014; 124:2921–2934. [PubMed: 24937432] 43. Cooley BC, Nevado J, Mellad J, et al. TGF-β Signaling Mediates Endothelial-to-Mesenchymal Transition (EndMT) During Vein Graft Remodeling. Sci Transl Med. 2014; 6:227ra234. 44. Hu MC, Shi M, Cho HJ, et al. Klotho and Phosphate Are Modulators of Pathologic Uremic Cardiac Remodeling. Journal of the American Society of Nephrology. 2015; 26:1290–1302. [PubMed: 25326585] 45. Towler DA, Bidder M, Latifi T, et al. Diet-induced diabetes activates an osteogenic gene regulatory program in the aortas of low density lipoprotein receptor-deficient mice. J Biol Chem. 1998; 273:30427–30434. [PubMed: 9804809] 46. Davies MR, Lund RJ, Mathew S, et al. Low turnover osteodystrophy and vascular calcification are amenable to skeletal anabolism in an animal model of chronic kidney disease and the metabolic syndrome. J Am Soc Nephrol. 2005; 16:917–928. [PubMed: 15743994] 47. Lund RJ, Davies MR, Brown AJ, et al. Successful treatment of an adynamic bone disorder with bone morphogenetic protein-7 in a renal ablation model. J Am Soc Nephrol. 2004; 15:359–369. [PubMed: 14747382] Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 12

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

48. Mathew S, Lund RJ, Chaudhary LR, et al. Vitamin D receptor activators can protect against vascular calcification. J Am Soc Nephrol. 2008; 19:1509–1519. [PubMed: 18448587] 49. Simone ND, Hall HA, Welt C, et al. Activin Regulates βA-Subunit and Activin Receptor Messenger Ribonucleic Acid and Cellular Proliferation in Activin-Responsive Testicular Tumor Cells. Endocrinology. 1998; 139:1147–1155. [PubMed: 9492049] 50. Liu ZH, Tsuchida K, Matsuzaki T, et al. Characterization of isoforms of activin receptorinteracting protein 2 that augment activin signaling. J Endocrinol. 2006; 189:409–421. [PubMed: 16648306] 51. Li L, Miano JM, Cserjesi P, et al. SM22α, a Marker of Adult Smooth Muscle, Is Expressed in Multiple Myogenic Lineages During Embryogenesis. Circ Res. 1996; 78:188–195. [PubMed: 8575061] 52. Abe YMT, Leung PC. Activin Receptor Signaling. Growth Factors (Chur, Switzerland). 2004; 22:105–110. 53. Langdon JM, Barkataki S, Berger AE, et al. RAP-011, an activin receptor ligand trap, increases hemoglobin concentration in hepcidin transgenic mice. Am J Hematol. 2015; 90:8–14. [PubMed: 25236856] 54. Mao J, Wang J, Liu B, et al. Low-density lipoprotein receptor-related protein-5 binds to axin and regulates the canonical Wnt signaling pathway. Molec Cell. 2001; 7:801–809. [PubMed: 11336703] 55. Lindberg K, Olauson H, Amin R, et al. Arterial klotho expression and FGF23 effects on vascular calcification and function. PLoS ONE. 2013; 8:e60658. [PubMed: 23577141] 56. Lim K, Lu T-S, Molostvov G, et al. Vascular Klotho Deficiency Potentiates the Development of Human Artery Calcification and Mediates Resistance to Fibroblast Growth Factor 23 / Clinical Perspective. Circulation. 2012; 125:2243–2255. [PubMed: 22492635] 57. Lindberg K, Amin R, Moe OW, et al. The Kidney Is the Principal Organ Mediating Klotho Effects. J Am Soc Nephrol. 2014; 25:2169–2175. [PubMed: 24854271] 58. Antsiferova M, Werner S. The bright and the dark sides of activin in wound healing and cancer. J Cell Sci. 2012; 125:3929–3937. [PubMed: 22991378] 59. Werner S, Alzheimer C. Roles of activin in tissue repair, fibrosis, and inflammatory disease. Cytokine & Growth Factor Reviews. 2006; 17:157–171. [PubMed: 16481210] 60. Yamashita S, Maeshima A, Kojima I, et al. Activin A Is a Potent Activator of Renal Interstitial Fibroblasts. J Am Soc Nephrol. 2004; 15:91–101. [PubMed: 14694161] 61. Maeshima A, Miya M, Mishima K, et al. Activin A: Autocrine Regulator of Kidney Development and Repair. Endocrine J. 2008; 55:1–9. [PubMed: 17827789] 62. Welt C, Sidis Y, Keutmann H, et al. Activins, Inhibins, and Follistatins: From Endocrinology to Signaling. A Paradigm for the New Millennium. Exp Biol Med. 2002; 227:724–752. 63. Sharpe RM, Turner KJ, McKinnell C, et al. Inhibin B Levels in Plasma of the Male Rat from Birth to Adulthood: Effect of Experimental Manipulation of Sertoli Cell Number. J Androl. 1999; 20:94–101. [PubMed: 10100479] 64. Yao Y, Zebboudj AF, Torres A, et al. Activin-like kinase receptor 1 (ALK1) in atherosclerotic lesions and vascular mesenchymal cells. Cardiov Res. 2007; 74:279–289. 65. Piek E, Heldin C-H, Ten Dijke P. Specificity, diversity, and regulation in TGF-β superfamily signaling. FASEB J. 1999; 13:2105–2124. [PubMed: 10593858] 66. Sun Y, Byon CH, Yuan K, et al. Smooth Muscle Cell–Specific Runx2 Deficiency Inhibits Vascular Calcification. Circ Res. 2012; 111:543–552. [PubMed: 22773442] 67. Speer MY, Li X, Hiremath PG, et al. Runx2/Cbfa1, but not loss of myocardin, is required for smooth muscle cell lineage reprogramming toward osteochondrogenesis. Journal of Cellular Biochemistry. 2010; 110:935–947. [PubMed: 20564193] 68. Matsuzaki T, Hanai S, Kishi H, et al. Regulation of Endocytosis of Activin Type II Receptors by a Novel PDZ Protein through Ral/Ral-binding Protein 1-dependent Pathway. Journal of Biological Chemistry. 2002; 277:19008–19018. [PubMed: 11882656] 69. Kurisaki A, Inoue I, Kurisaki K, et al. Activin induces long-lasting N-methyl-d-aspartate receptor activation via scaffolding PDZ protein activin receptor interacting protein 1. Neuroscience. 2008; 151:1225–1235. [PubMed: 18201830] Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 13

Author Manuscript Author Manuscript

70. Crease DJ, Dyson S, Gurdon JB. Cooperation between the activin and Wnt pathways in the spatial control of organizer gene expression. Proceedings of the National Academy of Sciences of the United States of America. 1998; 95:4398–4403. [PubMed: 9539748] 71. Gadue P, Huber TL, Paddison PJ, et al. Wnt and TGF-β signaling are required for the induction of an in vitro model of primitive streak formation using embryonic stem cells. Proc Natl Acad Sci USA. 2006; 103:16806–16811. [PubMed: 17077151] 72. Hansson M, Olesen DR, Peterslund JML, et al. A late requirement for Wnt and FGF signaling during activin-induced formation of foregut endoderm from mouse embryonic stem cells. Developmental Biology. 2009; 330:286–304. [PubMed: 19358838] 73. Xiao L, Yuan X, Sharkis SJ. Activin A Maintains Self-Renewal and Regulates Fibroblast Growth Factor, Wnt, and Bone Morphogenic Protein Pathways in Human Embryonic Stem Cells. Stem Cells. 2006; 24:1476–1486. [PubMed: 16456129] 74. Dussiot M, Maciel TT, Fricot A, et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in [beta]-thalassemia. Nat Med. 2014; 20:398–407. [PubMed: 24658077] 75. Lotinun S, Pearsall RS, Davies MV, et al. A soluble activin receptor Type IIA fusion protein (ACE-011) increases bone mass via a dual anabolic-antiresorptive effect in Cynomolgus monkeys. Bone. 2010; 46:1082–1088. [PubMed: 20080223] 76. Rheault MN, Kren SM, Thielen BK, et al. Mouse Model of X-Linked Alport Syndrome. J Am Soc Nephrol. 2004; 15:1466–1474. [PubMed: 15153557]

Author Manuscript Author Manuscript Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 14

Author Manuscript Author Manuscript Figure One.

Author Manuscript

Schematic of ActRIIA ligand trap and experimental design of its use in the ldlr−/− high fat fed CKD vascular calcification model. A, schematic diagram of the mouse fusion protein of the extracellular domain of ActRIIA and the Fc domain of IgG1 (RAP-011). B, experimental design of RAP-011 effects on the CKD-MBD in ldlr−/− high fat fed mice. Mice in four groups were fed the high fat diet beginning at 12 weeks (wks) of age. At 12 wks either sham operation (SO) or electrocautery cortical injury (EC) was performed. At 14 wks either SO or contralateral nephrectomy (NX) was performed. At 22 wks of life, vehicle treatment or RAP-011, 10mg/kg subcutaneous twice weekly, was instituted. WT, wild type mice on chow diet for normal reference levels. Sham, sham operated ldlr−/− high fat fed mice; CKD, CKD ldlr−/− high fat fed mice studied at 22 wks to establish levels of vascular calcium at the start of therapy; CKD V, CKD ldlr−/− high fat fed mice vehicle treated; CKD R, CKD ldlr−/− high fat fed mice RAP-011 treated; WT, Sham, CKD V and CKD R mice were euthanized at 28 weeks of age.

Author Manuscript Kidney Int. Author manuscript; available in PMC 2017 June 01.

Agapova et al.

Page 15

Author Manuscript Author Manuscript Figure Two.

Author Manuscript

Expression of ActRIIA in mouse aortas. A, Westerns for ActRIIA in aortic homogenates and immunoblot quantitation to the right. For the quantitation, n=4, **p

Ligand trap for the activin type IIA receptor protects against vascular disease and renal fibrosis in mice with chronic kidney disease.

The causes of cardiovascular mortality associated with chronic kidney disease (CKD) are partly attributed to the CKD-mineral bone disorder (CKD-MBD). ...
2MB Sizes 0 Downloads 6 Views