Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: J Mol Cell Cardiol. 2015 Dec 11;90:129–138. doi: 10.1016/j.yjmcc.2015.12.010

The TGF-β Pathway Mediates Doxorubicin Effects on Cardiac Endothelial Cells

Zuyue Sun c,1, Jill Schriewer a, Mingxin Tang b, Jerry Marlin a, Frederick Taylor c, Ralph V Shohet b, Eugene A Konorev a,*
PMCID: PMC4718883  NIHMSID: NIHMS747829  PMID: 26686989

Abstract

Elevated ALK4/5 ligands including TGF-β2 and activins have been linked to cardiovascular remodeling and heart failure. Doxorubicin (Dox) is commonly used as a model of cardiomyopathy, a condition that often precedes cardiovascular remodeling and heart failure. In 7-8-week-old C57Bl/6 male mice treated with Dox we found decreased capillary density, increased levels of ALK4/5 ligand and Smad2/3 transcripts, and increased expression of Smad2/3 transcriptional targets. Human cardiac microvascular endothelial cells (HCMVEC) treated with Dox also showed increased levels of ALK4/5 ligands, Smad2/3 transcriptional targets, a decrease in proliferation and suppression of vascular network formation in a HCMVEC and human cardiac fibroblasts co-culture assay. Our hypothesis is that the deleterious effects of Dox on endothelial cells are mediated in part by the activation of the TGF-β pathway. We used the inhibitor of ALK4/5 kinases SB431542 (SB) in concert with Dox to ascertain the role of TGF-β pathway activation in doxorubicin induced endothelial cell defects. SB prevented the suppression of HCMVEC proliferation in the presence of TGF-β2 and activin A, and alleviated the inhibition of HCMVEC proliferation by Dox. SB also prevented the suppression of vascular network formation in co-cultures of HCMVEC and human cardiac fibroblasts treated with Dox. Our results show that the inhibition of the TGF-β pathway alleviates the detrimental effects of Dox on endothelial cells in vitro.

Keywords: Cardiomyopathy, TGF-β, endothelial cells, angiogenesis, anthracyclines

1. Introduction

Patients with heart failure often suffer progressive deterioration of cardiac function. This may be due to increased hemodynamic stress leading to pathologic remodeling [1]. Cardiac remodeling is often considered to be a sign of an irreversible cardiac disease that is associated with a poor prognosis. Current treatments primarily slow the functional decline but cannot prevent or reverse the progressive nature of the disease.

A growing body of evidence links elevated levels of TGF-β superfamily ligands to the progression of heart failure. Specifically, increased cardiac levels of activin receptor-like kinase 4 (ALK41, ACVR1B) and ALK5 (TGFBR1) ligands TGF-β, activins and myostatin have been detected both in patients and animal models of heart failure [2-6]. The levels of these proteins are positively correlated with the severity of the condition [7, 8]. An ALK4/5 ligand binds to a cognate type II receptor, which then forms a complex with a type I receptor (Fig.1). As a result, the serine/threonine kinase activity of the type I receptor is activated to phosphorylate receptor-regulated Smad proteins (R-Smads) Smad2 and Smad3 [9]. By activating Smad2/3 signaling pathways, the ALK4/5 ligands regulate a broad range of processes including inflammation, tissue repair, cell proliferation and cell differentiation [9, 10].

Figure 1.

Figure 1

Scheme of ALK4/5 ligands and their receptors

In the cardiovascular system, TGF-β plays a major role in fibrotic cardiac remodeling. Expression of ALK4/5 ligands leads to the activation of fibroblasts in cardiac tissue by promoting myofibroblast differentiation, enhancing the synthesis of extracellular matrix proteins and regulating fibrotic remodeling of the diseased heart in a variety of conditions [11-14]. Inhibition of the TGF-β pathway alleviates left ventricular remodeling, systolic and diastolic dysfunction [15-17]. Although a key role for TGF-β in fibrotic cardiac remodeling has been established, its vascular effects have not been examined. ALK4/5 ligands affect multiple cell types regulating the growth and maintenance of vascular networks [18]. Recent studies show that endothelial cells treated with TGF-β ligands exhibit decreased proliferation, migration and angiogenesis [19-24]. Activin A has been identified as an inhibitor of angiogenesis in vivo [25, 26]. In order to understand the role of the TGF-β pathway in cardiac vascular remodeling, we tested the effects of a small molecular weight inhibitor of the TGF-β pathway on the formation of vascular networks by human cardiac microvascular endothelial cells.

In this study, we have utilized the exposure of mice and human cardiac endothelial cells to doxorubicin (Dox), which is commonly used as a model of cardiomyopathy and [27-29]. Clinical use of this effective anticancer agent has become restricted because of severe cardiac damage and mortality occurring in patients treated with this agent [30, 31]. Anthracycline antibiotics such as Dox cause a dose-related dilated cardiomyopathy when used to treat cancer patients [32]. Cancer survivors represent an important group of patients at risk of premature cardiovascular disease. The cardiovascular complications in former cancer patients are particularly difficult to treat [33]. Dox cardiomyopathy responds poorly to therapy and often progresses to fatal congestive heart failure [34]. Apoptosis of contractile cells in the heart, cardiomyocytes, has been implicated as a factor contributing to cardiac damage [35, 36]. However, adult cardiomyocytes may be resistant to doxorubicin-induced apoptosis [37, 38]. This study focuses on cardiac microvascular endothelial cells as a target of Dox in the heart.

Unlike other models of cardiomyopathy, Dox cardiomyopathy can be conveniently investigated by in vitro treatments of cardiac cells with Dox. We show that Dox causes profound vascular defects in cardiac tissue, which manifest as decreased capillary density in treated mouse hearts and suppressed formation of vascular networks by human cardiac endothelial cells in vitro. Vascular changes in the treated hearts occurred prior to the development of other manifestations of cardiomyopathy including cardiac fibrosis and deterioration of cardiac function. We detected the increased expression of TGF-β, related factors and their receptors in Dox treated mouse hearts and in cardiac cell cultures. The inhibition of ALK4/5 receptor kinase activity using a small molecular weight inhibitor enhanced vascular network formation by human cardiac endothelial cells treated with Dox, suggesting that this pathway could be a target for the treatment of cardiomyopathy.

2. Materials and Methods

2.1. Reagents and cell culture

Antibodies from Cell Signaling Technologies that were used for western blot and immunocytochemistry include anti-phospho Smad2 (clone 138D4), anti-Smad2 (clone D43B4), anti phospho Smad3 (C25A9), anti Smad3 (C67H9) and anti p21 (clone 12D1). Anti CD31 (Dako clone JC70A), anti Ki67 and anti-cardiac troponin I (Abcam polyclonal antibodies derived in rabbit) were used in immunostaining. Secondary antibodies to mouse and rabbit IgG (H+L) labeled with AlexaFluor 488 and 594 were used for immunocytochemistry (Life Technologies). Secondary antibodies to mouse and rabbit IgG (H+L) labeled with IRDye 800CW and 680LT (LI-COR Biosciences) were used in western blot. Biotinylated isolectin B4 and streptavidin-fluorescein were from Vector Laboratories.

Human cardiac microvascular endothelial cells (HCMVEC), human umbilical vein endothelial cells (HUVEC) and human cardiac fibroblasts (HCF) were purchased from Lonza. Cells were cultured in EGM2-MV, EGM2 and FGM3 complete media, respectively. All experiments were performed between passages six and seven. Cells were passaged using 0.05% Trypsin-EDTA that was neutralized with defined trypsin inhibitor (Life Technologies).

2.2. Doxorubicin treatments of mice

Seven-to-eight-week old male C57BL/6 mice were used in the study. Animals were bred and maintained at the Animal and Veterinary Service facility at the University of Hawaii JABSOM. All procedures described in this study were approved by the University of Hawaii IACUC and conformed to NIH guidelines for care and use of laboratory animals. A sterile solution of Dox in saline was administered intraperitoneally once a week at a dose of 3 mg/kg of body weight (n=24, with 6 Dox weekly injections per animal). Control mice were injected with saline (n=17). Echocardiography was performed after 6 weeks of Dox injections. Mice were weighed and sacrificed. The hearts were excised, weighed, and processed for histological and immunohistochemical analyses.

2.3. Histology and immunohistochemistry

Hearts were fixed by perfusion with IHC zinc fixative (BD Biosciences) for capillary staining, dehydrated in 30% sucrose in PBS overnight at 4°C and frozen-embedded in OCT compound. Cardiac sections were pre-incubated with permeabilization/blocking buffer (0.1% Triton X-100 and 3% BSA in PBS) for 30 min at room temperature, and then incubated sequentially with biotinylated isolectin B4 and streptavidin-fluorescein. The slides were cover-slipped in Fluoromount-G mounting medium (Southern Biotech) and examined by fluorescence microscopy using Zeiss LSM 510 confocal microscope. The number of capillaries in the field of view was quantified and normalized to the cardiomyocyte area (determined by staining with cardiac troponin I) using ImageJ software. For the collagen staining, hearts were fixed in 4% paraformaldehyde and stained at JABSOM Histology and Imaging core facility using Trichrome Staining Kit (Modified Gomori's) from ScyTek Laboratories (Logan, UT).

2.4. Endothelial cell proliferation and vascular network formation assays

Endothelial cell proliferation was measured by plating 1500 cells per well in 96 well plates. Beginning on day 0, one set of plates was fixed, nuclei were stained with Hoechst 33342 and the plate was imaged on the Cytation3 (Biotek). On days 0, 2, and 4 plates were treated according to the experimental protocol. On days 2, 4 and 6 plates were fixed and nuclei were similarly stained and imaged. The change in cell number or fold change are expressed as a change with respect to the cell counts made on day 0.

Vascular network formation assays were performed as described in [39] with modifications. Specifically, assays consisted of 2500 HCMVEC and 5000 HCF per well plated in 96 well plates using 100μl of endothelial cell media per well. Beginning on day 0, cells were treated every 48 hours according to experimental protocol. At the indicated day, cells were fixed and stained using mouse anti-CD31 and anti-mouse AlexaFluor 488 antibodies. Plates were imaged on the Cytation3 collecting overlapping images using the 4X objective. The images were stitched together using ImageJ and the stitched images analyzed for network length on the Metamorph software platform using the Angiogenesis module.

2.5. Immunocytochemistry

Cells were cultured in 96 well plates and treated according to the experimental protocol. At a given time point, cells were washed with PBS and fixed with 4% paraformaldehyde in PBS. Cells were washed, permeabilized with 0.1% Triton-X 100, blocked with 3% BSA in PBS, and stained overnight at 4°C with antibodies diluted in 3% BSA-PBS-0.05% Tween-20. Primary antibodies were detected with AlexaFluor labeled secondary antibodies diluted in 3% BSA-PBS-0.05% Tween-20. Nuclei were detected with 2 μM Hoechst 33342 in PBS-0.05% Tween-20. Plates were imaged using the Cytation3 (Biotek). The nuclei positive for Hoechst 33342 were counted. The number of nuclei positive for a specific protein was determined using Gen5 Image Plus software.

2.6. Immunoblotting and ELISA

Cells were cultured and treated according to experimental protocol. At a given time point, culture dishes were placed on ice and washed two times with ice cold PBS. Cells were lysed using M-PER (Pierce) with the HALT inhibitor cocktail (Pierce) on ice for 15 minutes. Cell lysates were scraped into the extraction buffer and the insoluble fraction was pelleted at 12,000g for 10 minutes at 2°C. The lysate was assayed for protein concentration (BCA assay, Pierce). Samples were denatured and reduced (4X sample buffer with β-mercaptoethanol, Bio-Rad). The resolved protein was transferred to low fluorescent PVDF using the Biorad Turboblot system. Membranes were blocked with a SeaBlock blocking buffer (ThermoFisher). Primary antibodies were diluted in blocking buffer with 0.1% Tween-20 and incubated overnight at 4°C. Secondary antibodies with the fluorescent tags 800CW and 680LT were diluted in blocking buffer with 0.1% Tween-20 and incubated one hour at room temperature. Membranes were imaged on the Odyssey imaging system (LI-COR Biosciences) using Image Studio software for image capture and analysis. The amount of phosphorylated Smad2 and Smad3 per lane was normalized to the total parent proteins. The amount of total Smad2/3 proteins per lane was normalized to β-actin.

Supernatants of cell culture experiments were harvested for measurements of TGF-β1, TGF-β2 and activin A production. Supernatants were clarified by centrifugation at 10,000g for 10 minutes at 4°C and then stored at −80°C. Concentrations of ligands were measured using Quantikine ELISA kits (R&D Systems). Cells were lysed using M-PER extraction reagent (Pierce) per manufacturer's protocol and the protein concentration determined. The amount of ligand secreted in the culture media was normalized to the protein concentration in lysed cells.

2.7. Quantitative PCR and PCR array assays

Co-cultures were conducted using trans-well inserts. 95,000 fibroblasts per insert and 115,000 endothelial cells per well were plated in a 6 well plate format (Corning/Costar). Both the inserts and wells were treated according to experimental protocol. Cultured cells were lysed with TRIzol and the RNA extracted with PureLink Mini Kit (Life Technologies). cDNA was generated (High Fidelity RNA to cDNA, Life Technologies) and real time PCR was performed using human TaqMan primer probe sets (Life Technologies) that span two exons. The threshold counts were normalized to the relative abundance of HPRT1 using the comparative CT method. Fold changes in gene expression are calculated as the fold change versus control.

Freshly harvested cardiac tissues from control and Dox treated mice used in PCR array assay were quickly immersed into RNAlater RNA stabilization reagent and homogenized in TRIzol. TURBO DNA-free Kit (Life Technologies) was used to remove traces of DNA from preparations of total RNA. Gene expression in control and Dox treated hearts was examined using mouse TGF-β/BMP Signaling Pathway RT2 Profiler PCR Array (QIAGEN).

2.8. Statistical analysis

Results are expressed as mean ± Standard Error of Mean (SEM). Statistical analysis was performed initially using normality test. Normally distributed data were subjected to parametric tests, such as t-test or analysis of variance. We used one way or two way analysis of variance to compare more than two experimental groups within a data set. Significance was accepted at the p<0.05 level.

3. Results

3.1. Microvascular defects in doxorubicin treated hearts

Mice received weekly injections of Dox or saline for 6 consecutive weeks, and hearts were excised at the end of the treatment course for further analyses. Specifically, reduced capillary density was detected in hearts of mice treated with Dox (Fig. 2A and B). Moderately decreased heart weight-to-body weight (HW/BW) ratio was also detected in Dox treated hearts (Fig. 2C). We performed analysis of gene expression in mouse hearts after 6 weeks of Dox treatment utilizing mouse BMP/TGF-β pathway PCR array. A partial list of the transcripts that were ≥2-fold more abundant in Dox-treated hearts is presented in Table 1. This group included ALK4/5 ligands and their receptors (Acvr2a, Inhba, Tgfb1, Tgfbr1), transcription factors that function in the canonical TGF-β/activin pathway (Smad2, Smad3), known TGF-β target genes (Serpine1, Thbs1, and Cdkn1b), and proteins that participate in storing and/or activation of TGF-β (Ltbp1, Thbs1). Complete lists of up-, downregulated and unchanged by Dox transcripts is presented in Supplementary Table 1. Thus, we detected changes in transcript abundance that are indicative of the increased activity of TGF-β pathway, with an associated decrease in capillary density in Dox treated mouse hearts.

Figure 2. Microvascular defects in doxorubicin treated mouse hearts.

Figure 2

Mice received weekly intraperitoneal injections of Dox (3 mg/kg) or saline for six weeks. (A) Cardiac tissue sections were stained for capillaries using isolectin B4 (IL-B4, green) and counterstained for cardiac troponin I (blue). (B) Quantitative analysis of capillary density is presented as number of capillaries per field of view normalized to myocardial (cardiac troponin I positive) area (n=3 hearts for saline group; n=3 hearts for Dox treated group). (C) Heart weight (HW) and body weight (BW) were taken at the end of the Dox treatment period to determine HW/BW ratio (n=3 for saline group; n=4 for Dox treated group). *p<0.05 as compared with control. Scale bar, 100 μm.

Table 1.

TGFβ/activin pathway genes upregulated in doxorubicin treated mouse hearts

Gene Symbol Gene Full Name
Acvr2a activin A receptor, type IIA
Cdkn1b cyclin-dependent kinase inhibitor 1B
Inha inhibin, alpha
Inhba inhibin, beta A
Ltbp1 latent transforming growth factor beta binding protein 1
Serpine1 serine (or cysteine) peptidase inhibitor, clade E, member 1
Smad2 SMAD family member 2
Smad3 SMAD family member 3
Smurf1 SMAD specific E3 ubiquitin protein ligase 1
Tgfb1 transforming growth factor, beta 1
Tgfbr1 transforming growth factor, beta receptor 1
Thbs1 thrombospondin 1

3.2. TGF-β pathway inhibitor reduces production of ALK4/5 ligands in doxorubicin treated cardiac cells

Since our in vivo experiments suggested an increased activity of TGF-β/activin pathway in Dox treated hearts, we examined the effects of Dox on the production of these factors by human cardiac cells. The production of activin A and TGF-β isoforms was increased in Dox treated HCMVEC and their co-cultures with cardiac fibroblasts (Supplementary Fig. 1). Furthermore, Dox increased Smad2/3 translocation in HCMVEC by approximately 2-fold and augmented Smad2/3 translocation induced by TGF-β2 in HCMVEC (Fig.3A and B). In order to assess gene expression in co-culture experiments, we utilized tissue culture inserts with HCMVEC and HCF plated in lower and upper chambers, respectively. Specifically, we detected increased expression of genes encoding ALK4/5 ligands and their receptors (Inhba, TGFB1, TGFB2, TGFBR1) as well as Smad2/3 transcriptional target plasminogen activator inhibitor-1 (SERPINE1) in Dox-treated HCMVEC co-cultured with HCF using trans-well inserts(Fig. 3C).

Figure 3. Enhancement of Smad2/3 nuclear localization and TGF-β pathway by Dox.

Figure 3

(A) HCMVEC cultures were treated with 16 nM Dox for 72 hours. Cultures were starved for six hours, then treated with 0.3 ng/ml TGF-β2 and stained for Smad2/3 and CD31. (B) Smad2/3 nuclear localization measured as the percentage of nuclei that are positive for Smad2/3. (C) HCMVEC and HCF were plated in the lower and upper wells of a tissue culture insert, respectively, and treated with 32 nM Dox and 1 μM SB431542 (SB) for 48 hours. Gene expression in HCMVEC was examined using quantitative PCR and TaqMan gene expression assays. Expression of HPRT1 gene was used as an endogenous control. (D) HCMVEC and HCF were plated in a mixed co-culture and treated with 32 nM Dox and 1 μM SB for 72 hours. Concentrations of TGF-β1 and activin A proteins in culture media were examined using ELISA and normalized to total cell protein. * and #, p<0.05 as compared with control and Dox groups, respectively. Scale bar, 100μM.

We tested the efficacy of ALK4/5/7 receptor kinase inhibitor SB431542 (SB) in preventing effects of TGF-β pathway activation in endothelial cells. SB blocked Smad2/3 phosphorylation and their nuclear translocation in HCMVEC treated with TGF-β2 and activin A (Fig. 4 and Supplementary Fig. 2) indicating that this pathway is functional in cardiac endothelial cells and inhibited by SB. Importantly, SB significantly decreased production of activin A and TGF-β1 proteins in Dox-treated co-cultures.

Figure 4. Inhibition of Smad2/3 phosphorylation and nuclear translocation in HCMVEC by SB431542.

Figure 4

(A) HCMVEC were cultured in complete EGM2-MV media, starved overnight, treated with 3 ng/ml TGF-β2 for 60 minutes and processed for immunoblotting. SB431542 (SB) was added to HCMVEC cultures 60 min prior to TGF-β2 treatment. (B) HCMVEC were treated with TGF-β2 and SB as described in (A) and stained for Smad3 (red). Nuclei were counterstained with Hoechst 33342 (blue). (C) Statistical analysis of Smad3 nuclear translocation images. * and #, p<0.05 as compared with control and TGF-β2, respectively. Scale bar, 50 μm.

Similarly, TGF-β pathway inhibitor reversed upregulated expression of genes in HCMVEC that function in TGF-β/activin pathway (Fig. 3C and D). Thus, studies utilizing HCMVEC and their co-culture with cardiac fibroblasts corroborated our in vivo findings regarding upregulated production of ALK4/5 ligands in Dox treated mouse hearts

3.3. TGF-β pathway inhibitor enhances proliferation of human endothelial cells treated with ALK4/5 ligands and doxorubicin

Since our results provided evidence for the increased production of ALK4/5 ligands by Dox treated cardiac cells, we tested effects of TGF-β2 and activin A on HCMVEC proliferation (Fig. 5). Both of these ligands severely reduced both the numbers of HCMVEC over 6-day treatment period and percentage of cells positive for the proliferation marker Ki67 (p<0.05). Treatment with SB increased numbers of both untreated and ligand treated HCMVEC and prevented the decrease in Ki67-positive cells that were treated with TGF-β2 and activin A.

Figure 5. TGF-β pathway inhibitor enhances proliferation of HCMVEC treated with ALK4/5 ligands.

Figure 5

HCMVEC were treated with 10 ng/ml activin A (A), 0.3 ng/ml TGF-β2 (B) and nuclei were stained at indicated time points with Hoechst 33342 for the total cell number evaluation. SB431542 (SB) was present at 1 μM concentration. * and #, p<0.05 as compared to control and ligand only treated groups, respectively. (C, D) HCMVEC cultures were treated with Dox and SB as described and stained for CD31 (green) and Ki67 (red). Images were acquired and analyzed using Cytation 3 imaging system. * p<0.05 for SB(+) groups as compared to the corresponding SB(−) groups. Scale bar, 100 μm.

In separate experiments, we examined the proliferation of HCMVEC treated with low (4-16 nM) concentrations of Dox. Dox treatment decreased numbers of endothelial cells in a concentration-dependent manner (Fig. 6A). We also detected upregulated expression of p21WAF1/CIP1 protein and reduced expression of Ki67 in treated cells (Fig. 6C and D). In order to examine if inhibition of proliferation may be due to the production of ALK4/5 ligands by Dox treated endothelial cells, we utilized SB in these experiments. Indeed, SB increased numbers of HCMVEC in all Dox treatment groups suggesting that activation of TGF-β pathway may have contributed to the inhibition of endothelial cell proliferation in this setting (Fig. 6B).

Figure 6. Doxorubicin inhibits proliferation of HCMVEC.

Figure 6

(A) HCMVEC were treated with the indicated concentrations of Dox and nuclei were stained with Hoechst 33342 for the total cell counts at the indicated time points. *, p<0.05 as compared with control. (B) HCMVEC were treated with the indicated concentrations of Dox and 1 μM SB431542 (SB) for 6 days. Nuclei were stained with Hoechst 33342 for the total cell counts per well. *, p<0.05 as compared to SB(−) group. (C and D) HCMVEC were treated with the indicated concentrations of Dox and stained for CD31 (green) and Ki67 or p21 (red). Images were acquired and analyzed using Cytation 3 imaging system. *, p<0.05 as compared with control. Scale bar, 100 μm.

3.4. TGF-β pathway inhibitor enhances formation of vascular networks by doxorubicin treated human endothelial cells

Microvascular defects observed in cardiomyopathic hearts of Dox-treated mice prompted us to examine the direct action of this agent on vascular network formation. We utilized co-culture systems of HCMVEC or HUVEC with human cardiac fibroblasts. These systems develop intricate vessel-like structures after 5 to 7 days in co-culture (Fig. 7A). Doxorubicin potently inhibited the formation of vascular networks in these co-culture systems. Quantitative evaluation of captured images demonstrated that Dox suppressed vascular network formation and reduced the number of endothelial cells in co-cultures by 70% and 85%, respectively (Fig. 7A and B). Similar results were obtained with HUVEC/HCF co-cultures (data not shown). Further analysis has shown that Dox did not cause acute endothelial cell toxicity or apoptotic cell death within the range of concentrations used in these experiments, but caused severe inhibition of HCMVEC proliferation in treated co-cultures (data not shown). SB enhanced vascular network formation in HCMVEC/HCF co-cultures treated with Dox (Fig. 7A and B). Similarly, endothelial cell number was significantly increased by SB in both intact and Dox treated co-cultures (Fig. 7C). Similar results were obtained with a structurally different ALK4/5 inhibitor SB525334 (data not shown). It is important to note that SB did not increase numbers of fibroblasts in both control and Dox treated co-cultures (Fig. 7D). Thus, Dox suppressed formation of vessel-like networks in HCMVEC/HCF co-cultures, and its inhibitory effect was alleviated by the TGF-β pathway inhibitor.

Figure 7. TGF-β pathway inhibitor enhances formation of vessel-like structures by doxorubicin treated HCMVEC.

Figure 7

(A) HCMVEC/HCF co-cultures (2,500 HCMVEC and 5,000 HCF) were treated with 32 nM Dox and 1 μM SB431542 (SB), and stained at day 7 for CD31 to visualize the vessel-like networks. (B) Day 7 network images were analyzed using Metamorph Angiogenesis module software and presented as total length of vessel-like structures per well. (C and D) Day 3 co-cultures were stained for CD31 and Hoechst 33342 to visualize HCMVEC and HCF. Numbers of endothelial cells and fibroblasts per well were quantified using Gen5 Image Plus software. * and #, p<0.05 as compared to untreated control and Dox only treated groups, respectively. Scale bar, 500 μm.

4. Discussion

Dox caused microvascular defects in the hearts of treated mice and suppression of the formation of vessel-like structures in HCMVEC co-cultured with HCF. We detected changes in gene expression that were indicative of the increased activity of the TGF-β pathway. Incubation of HCMVEC with either TGF-β2 or Activin A inhibited proliferation while a TGF-β pathway inhibitor abolished the defect in endothelial cells. Similarly, Dox inhibited HCMVEC proliferation but a TGF-β inhibitor alleviated this effect of Dox and enhanced formation of vessel-like structures in Dox treated co-cultures.

Decreased cardiac capillary density is considered to be a sign of microvascular deficit [40], which, in the case of Dox treatment, developed before other known markers of Dox cardiomyopathy including depressed cardiac function and myocardial fibrosis (data not shown). Reduced capillary density preceded the development of tissue hypoxia in renal cortex of Dox treated animals [41]. Microvascular deficit induced by the trapping of VEGF in myocardial tissue has been shown to trigger cardiac remodeling and failure while restoring VEGF levels reversed remodeling and rescued compromised ventricular function [40]. Microvascular deficiency is a crucial factor that causes hypertrophic, ischemic and cardiomyopathic conditions to progress towards a decompensated functional state. Strategies aimed at the activation of angiogenesis delay or prevent decompensation of cardiac function [42-46]. Thus, our data and literature considerations suggest that endothelial defects caused by Dox in the heart may contribute to the microvascular deficiency and development of cardiomyopathy in treated hearts.

4.1. Endothelial cell as a target of doxorubicin in the heart

Several cardiac cell types, including cardiomyocytes, cardiac progenitor cells, and endothelial cells have been considered as targets of doxorubicin in the heart that contribute to the development of cardiomyopathy [28, 35, 37, 47]. Huang et al. [28] have previously reported decreased capillary density in hearts treated with Dox. Interestingly, a study performed on hair-follicle vascular network found that endothelial cells were much more vulnerable to the damaging effects of Dox than hair follicle stem cells [48].

Apoptosis was considered to be the primary mechanism of endothelial injury by Dox [49, 50]. Indeed, our previous study supported this mechanism of Dox induced endothelial cell injury [51]. Concentrations of Dox used in these earlier studies and many other studies induced a strong apoptotic response that resulted in death of most endothelial cells in culture within several days. Such an apparent acute endothelial cell injury does not occur in patients treated with Dox [32]. Indeed, exposure of patients to anthracyclines often results in delayed cardiovascular toxicity, which manifests itself as left ventricular dysfunction that ultimately progresses to congestive heart failure [32, 33, 52, 53]. In addition, clinical follow-up studies detected chronic vascular inflammation and early atherosclerotic lesions in survivors of Hodgkin lymphoma and osteosarcoma treated with Dox and other chemotherapeutic agents [54, 55].

In the current study we used concentrations of Dox that were comparable to its plasma levels in animals after the injection of this agent [56], and did not detect apoptotic or necrotic endothelial cell death markers (data not shown). Our earlier studies with Dox showed that it is avidly and rapidly taken up by the cultured cells [57]. In fact, concentration of Dox in cultured cells typically exceeds its levels in culture media by 50 to 100-fold [58]. Estimated cellular concentrations of Dox in our in vitro experiments are similar to cardiac levels of this agent in animals received a clinically relevant dose of Dox [59-61]. Thus, we believe that the present study was conducted using Dox concentrations that are commonly obtained during clinical use of the agent, and the types of endothelial defects observed at these concentrations of Dox may therefore be clinically relevant.

Another previously suggested mechanism of endothelial damage by Dox involves either decreased expression of VEGF by treated tissues or VEGF receptor 2 (VEGFR2) by exposed endothelial cells [62-64]. At the concentrations that were used in the current study, Dox did not significantly change production of VEGF by HCMVEC/HCF co-cultures or expression of VEGFR2 by HCMVEC (data not shown). The endothelial cell growth media used in our experiments with mono-cultures and co-cultures was supplemented with human recombinant VEGF. We show that Dox significantly decreased numbers of endothelial cells, both in mono-cultures and co-cultures, as a result of inhibition of proliferation in the presence of VEGF. Specifically, it decreased expression of Ki67, a cell proliferation marker, and increased abundance of p21WAF1/CIP1 protein in treated endothelial cells. Induction of p21WAF1/CIP1 and cell cycle arrest are known consequences of the p53 transactivation program. Dox has been previously shown by us and others to induce transcriptional activation of p53 in endothelial cells [49, 65]. However, Dox caused neither p53 transactivation nor upregulation of p53 target genes in HCMVEC at low nanomolar concentrations used in the current study (data not shown).

4.2. Endothelial defects due to the increased activity of TGF-β pathway

Our initial observation of increased expression of genes that function in TGF-β pathway in Dox treated hearts was supported by our in-vitro experiments with human cardiac cell types. Specifically, we found that Dox increased production of ALK4/5 ligands by co-cultures of HCMVEC/HCF, and increased expression of ALK4/5 ligands and known TGF-β target genes. It has been known that TGF-β inhibits proliferation of cells of epithelial origin [66, 67]. We show in this study that ALK4/5 ligands, TGF-β2 and activin A, suppress proliferation of human cardiac endothelial cells. Several studies have previously reported inhibition of proliferation and migration by ALK4/5 ligands in endothelial cells of other origins [19, 20, 24]. Therefore, it is plausible that inhibition of ALK4/5 receptor signaling will enhance formation and improve maintenance of microvascular networks via direct effects on endothelial cell proliferation and migration. Indeed, suppression of TGF-β signaling by ALK4/5/7 receptor kinases inhibitor SB431542 [68] in endothelial cells led to the increased vascular sprouting by immortalized mouse endothelial cells and HUVEC in vitro [23, 69], preserved microvascular networks in such conditions as radiation injury and chronic kidney disease [70, 71] and increased proliferation of human embryonic stem cell derived endothelial cells [72, 73]. Likewise, SB431542 facilitated direct conversion of embryonic and adult cardiac fibroblasts into induced cardiomyocytes [74]. Increased efficiency of the fibroblast-to-cardiomyocyte reprogramming by SB431542 may potentially enhance regenerative capacity of cardiac muscle.

Effects of inhibition of TGF-β signaling in cardiac endothelial cells have not been reported before. We show here that SB431542 prevents the suppression of HCMVEC proliferation caused by ALK4/5 ligands. In addition, this inhibitor enhanced formation of vessel-like structures in control co-cultures and alleviated the inhibition of proliferation as well as suppression of formation of vessel-like networks by HCMVEC induced by Dox. These effects are likely due to the blocking the effects of ALK4/5 ligands on endothelial cells in both control and Dox treated co-cultures. In a co-culture, endothelial cells are exposed to higher concentrations of TGF-β and activin that are produced by fibroblasts, and Dox further increases production of these factors in this system. Increased levels of ALK4/5 ligands contribute to the suppression of endothelial cell proliferation in co-culture while SB431542 blocks their effects on endothelial cells to enhance their proliferation and vascular network formation. Importantly, in addition to the inhibition of signaling through ALK4/5 receptors by preventing Smad2/3 phosphorylation and nuclear translocation, SB decreased production of activin A and TGF-β1 by human cardiac cells. This effect on synthesis may further inhibit TGF-β pathway activity, interrupting the auto-induction loop whereby TGF-β1 and activins enhance their own expression [75-78].

Interestingly, SB431542 increased proliferation of endothelial cells but decreased numbers of fibroblasts in control co-culture experiments. Indeed, TGF-β is known to increase proliferation of fibroblastic cells and cancer cells that have inactivating mutations in the canonical TGF-β pathway [79-82]. TGF-β may contribute to the tumor growth and progression by enhancing fibroblast proliferation and their migration of into the tumor [83]. Cancer associated fibroblasts produce increased amounts of growth factors and cytokines that enhance both proliferation of tumor cells and angiogenesis, and increase resistance of cancer cells to chemotherapeutic agents [84]. In addition, TGF-β ligands are potent inducers of the epithelial-to-mesenchymal transition in transformed cells, leading to their enhanced migration and invasiveness [85]. Inhibition of TGF-β pathway with SB431542 sensitized resistant cancer cells to chemotherapy and suppressed cancer cell proliferation, migration and metastases [86, 87]. Thus, using SB431542 or other TGF-β pathway inhibitor under such circumstances to supplement the chemotherapy with Dox may not only enhance its anticancer efficiency but also protect the heart from the Dox toxicity.

4.3. Conclusions

Small molecular weight inhibitors of TGF-β pathway have been reported to alleviate fibrosis [88] and peritubular capillary defects [89] in Dox induced nephropathy. The results of the present study suggest that Dox causes increased expression of ALK4/5 ligands and vascular remodeling in treated mouse hearts, with reduced myocardial capillary density. This type of microvascular remodeling has been described in other types of cardiomyopathies, including diabetic and idiopathic dilated cardiomyopathies [43, 90]. Myocardial microvascular deficiency promotes the progression of cardiac disease. Results of this study suggest inhibition of TGF-β pathway as a plausible strategy aimed at the preservation and enhancement of myocardial capillary networks and prevention of microvascular remodeling.

Supplementary Material

PCR array
Suppl. Fig. 1
Suppl. Fig. 2

Highlights.

  • Doxorubicin causes decreased capillary density in treated mouse hearts

  • Doxorubicin increases expression of ALK4/5 ligands in cardiac cells

  • ALK4/5 ligands suppress cardiac endothelial cell proliferation

  • Doxorubicin inhibits cardiac endothelial cells proliferation and angiogenesis

  • ALK4/5/7 kinase inhibitor alleviates the anti-angiogenic effect of doxorubicin

ACKNOWLEDGEMENTS

This work was supported in part by National Institutes of Health 2P20RR016467-09A1 INBRE II grant. The funding source had no involvement in study design, the collection, analysis and interpretation of data, the writing of the report, and the decision to submit the article for publication.

Footnotes

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.

1

ABBREVIATIONS: HCMVEC, human cardiac microvascular endothelial cells; HUVEC, human umbilical vein endothelial cells; HCF, human cardiac fibroblasts; Dox, doxorubicin; ALK4/5, activin receptor-like kinase 4/5.

The authors declare no conflicts of interests in the publication of this manuscript.

REFERENCES

  • 1.Heusch G, Libby P, Gersh B, Yellon D, Bohm M, Lopaschuk G, et al. Cardiovascular remodelling in coronary artery disease and heart failure. Lancet. 2014;12:60107–0. doi: 10.1016/S0140-6736(14)60107-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fukushima N, Matsuura K, Akazawa H, Honda A, Nagai T, Takahashi T, et al. A crucial role of activin A-mediated growth hormone suppression in mouse and human heart failure. PLoS One. 2011;6:e27901. doi: 10.1371/journal.pone.0027901. doi: 10.1371/journal.pone.0027901. Epub 2011 Dec 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Oshima Y, Ouchi N, Shimano M, Pimentel DR, Papanicolaou KN, Panse KD, et al. Activin A and follistatin-like 3 determine the susceptibility of heart to ischemic injury. Circulation. 2009;120:1606–15. doi: 10.1161/CIRCULATIONAHA.109.872200. doi: 10.161/CIRCULATIONAHA.109.872200. Epub 2009 Oct 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Baan J, Varga ZV, Leszek P, Ku Mierczyk M, Baranyai T, Dux L, et al. Myostatin and IGF-I signaling in end-stage human heart failure: a qRT-PCR study. J Transl Med. 2015;13:1. doi: 10.1186/s12967-014-0365-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mahmoudabady M, Mathieu M, Dewachter L, Hadad I, Ray L, Jespers P, et al. Activin-A, transforming growth factor-beta, and myostatin signaling pathway in experimental dilated cardiomyopathy. J Card Fail. 2008;14:703–9. doi: 10.1016/j.cardfail.2008.05.003. doi: 10.1016/j.cardfail.2008.05.003. Epub Jun 11. [DOI] [PubMed] [Google Scholar]
  • 6.Khan S, Joyce J, Margulies KB, Tsuda T. Enhanced bioactive myocardial transforming growth factor-beta in advanced human heart failure. Circ J. 2014;78:2711–8. doi: 10.1253/circj.cj-14-0511. Epub 014 Oct 9. [DOI] [PubMed] [Google Scholar]
  • 7.Gruson D, Ahn SA, Ketelslegers JM, Rousseau MF. Increased plasma myostatin in heart failure. Eur J Heart Fail. 2011;13:734–6. doi: 10.1093/eurjhf/hfr024. doi: 10.1093/eurjhf/hfr024. Epub 2011 Apr 4. [DOI] [PubMed] [Google Scholar]
  • 8.Yndestad A, Ueland T, Oie E, Florholmen G, Halvorsen B, Attramadal H, et al. Elevated levels of activin A in heart failure: potential role in myocardial remodeling. Circulation. 2004;109:1379–85. doi: 10.1161/01.CIR.0000120704.97934.41. Epub 2004 Mar 1. [DOI] [PubMed] [Google Scholar]
  • 9.Massague J, Seoane J, Wotton D. Smad transcription factors. Genes Dev. 2005;19:2783–810. doi: 10.1101/gad.1350705. [DOI] [PubMed] [Google Scholar]
  • 10.Dennler S, Itoh S, Vivien D, ten Dijke P, Huet S, Gauthier JM. Direct binding of Smad3 and Smad4 to critical TGF beta-inducible elements in the promoter of human plasminogen activator inhibitor-type 1 gene. Embo J. 1998;17:3091–100. doi: 10.1093/emboj/17.11.3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ieronimakis N, Hays AL, Janebodin K, Mahoney WM, Jr., Duffield JS, Majesky MW, et al. Coronary adventitial cells are linked to perivascular cardiac fibrosis via TGFbeta1 signaling in the mdx mouse model of Duchenne muscular dystrophy. J Mol Cell Cardiol. 2013;63:122–34. doi: 10.1016/j.yjmcc.2013.07.014. 10.1016/j.yjmcc.2013.07.014. Epub Aug 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Westermann D, Lindner D, Kasner M, Zietsch C, Savvatis K, Escher F, et al. Cardiac inflammation contributes to changes in the extracellular matrix in patients with heart failure and normal ejection fraction. Circ Heart Fail. 2011;4:44–52. doi: 10.1161/CIRCHEARTFAILURE.109.931451. doi: 10.1161/CIRCHEARTFAILURE.109.931451. Epub 2010 Nov 12. [DOI] [PubMed] [Google Scholar]
  • 13.McNally EM, Goldstein JA. Interplay between heart and skeletal muscle disease in heart failure: the 2011 George E. Brown Memorial Lecture. Circ Res. 2012;110:749–54. doi: 10.1161/CIRCRESAHA.111.256776. doi: 10.1161/CIRCRESAHA.111.256776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kapur NK, Wilson S, Yunis AA, Qiao X, Mackey E, Paruchuri V, et al. Reduced endoglin activity limits cardiac fibrosis and improves survival in heart failure. Circulation. 2012;125:2728–38. doi: 10.1161/CIRCULATIONAHA.111.080002. doi: 10.1161/CIRCULATIONAHA.111.080002. Epub 2012 May 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Villar AV, Garcia R, Llano M, Cobo M, Merino D, Lantero A, et al. BAMBI (BMP and activin membrane-bound inhibitor) protects the murine heart from pressure-overload biomechanical stress by restraining TGF-beta signaling. Biochim Biophys Acta. 2013;1832:323–35. doi: 10.1016/j.bbadis.2012.11.007. doi: 10.1016/j.bbadis.2012.11.007. Epub Nov 17. [DOI] [PubMed] [Google Scholar]
  • 16.Tan SM, Zhang Y, Connelly KA, Gilbert RE, Kelly DJ. Targeted inhibition of activin receptor-like kinase 5 signaling attenuates cardiac dysfunction following myocardial infarction. Am J Physiol Heart Circ Physiol. 2010;298:H1415–25. doi: 10.1152/ajpheart.01048.2009. doi: 10.152/ajpheart.01048.2009. Epub 2010 Feb 12. [DOI] [PubMed] [Google Scholar]
  • 17.Kuwahara F, Kai H, Tokuda K, Kai M, Takeshita A, Egashira K, et al. Transforming growth factor-beta function blocking prevents myocardial fibrosis and diastolic dysfunction in pressure-overloaded rats. Circulation. 2002;106:130–5. doi: 10.1161/01.cir.0000020689.12472.e0. [DOI] [PubMed] [Google Scholar]
  • 18.Nakagawa T, Li JH, Garcia G, Mu W, Piek E, Bottinger EP, et al. TGF-beta induces proangiogenic and antiangiogenic factors via parallel but distinct Smad pathways. Kidney Int. 2004;66:605–13. doi: 10.1111/j.1523-1755.2004.00780.x. [DOI] [PubMed] [Google Scholar]
  • 19.McCarthy SA, Bicknell R. Inhibition of vascular endothelial cell growth by activin-A. J Biol Chem. 1993;268:23066–71. [PubMed] [Google Scholar]
  • 20.Maroni D, Davis JS. TGFB1 disrupts the angiogenic potential of microvascular endothelial cells of the corpus luteum. J Cell Sci. 2011;124:2501–10. doi: 10.1242/jcs.084558. doi: 10.1242/jcs.084558. Epub 2011 Jun 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zerlanko BJ, Bartholin L, Melhuish TA, Wotton D. Premature senescence and increased TGFbeta signaling in the absence of Tgif1. PLoS One. 2012;7:e35460. doi: 10.1371/journal.pone.0035460. doi: 10.1371/journal.pone.0035460. Epub 2012 Apr 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Goumans MJ, Valdimarsdottir G, Itoh S, Rosendahl A, Sideras P, ten Dijke P. Balancing the activation state of the endothelium via two distinct TGF-beta type I receptors. Embo J. 2002;21:1743–53. doi: 10.1093/emboj/21.7.1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Arnold TD, Niaudet C, Pang MF, Siegenthaler J, Gaengel K, Jung B, et al. Excessive vascular sprouting underlies cerebral hemorrhage in mice lacking alphaVbeta8-TGFbeta signaling in the brain. Development. 2014;141:4489–99. doi: 10.1242/dev.107193. doi: 10.1242/dev.107193. Epub 2014 Nov 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Castanares C, Redondo-Horcajo M, Magan-Marchal N, ten Dijke P, Lamas S, Rodriguez-Pascual F. Signaling by ALK5 mediates TGF-beta-induced ET-1 expression in endothelial cells: a role for migration and proliferation. J Cell Sci. 2007;120:1256–66. doi: 10.1242/jcs.03419. [DOI] [PubMed] [Google Scholar]
  • 25.Kaneda H, Arao T, Matsumoto K, De Velasco MA, Tamura D, Aomatsu K, et al. Activin A inhibits vascular endothelial cell growth and suppresses tumour angiogenesis in gastric cancer. Br J Cancer. 2011;105:1210–7. doi: 10.1038/bjc.2011.348. doi: 10.038/bjc.2011.348. Epub Sep 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Breit S, Ashman K, Wilting J, Rossler J, Hatzi E, Fotsis T, et al. The N-myc oncogene in human neuroblastoma cells: down-regulation of an angiogenesis inhibitor identified as activin A. Cancer Res. 2000;60:4596–601. [PubMed] [Google Scholar]
  • 27.Houser SR, Margulies KB, Murphy AM, Spinale FG, Francis GS, Prabhu SD, et al. Animal models of heart failure: a scientific statement from the American Heart Association. Circ Res. 2012;111:131–50. doi: 10.1161/RES.0b013e3182582523. doi: 10.1161/RES.0b013e3182582523. Epub 2012 May 17. [DOI] [PubMed] [Google Scholar]
  • 28.Huang C, Zhang X, Ramil JM, Rikka S, Kim L, Lee Y, et al. Juvenile exposure to anthracyclines impairs cardiac progenitor cell function and vascularization resulting in greater susceptibility to stress-induced myocardial injury in adult mice. Circulation. 2010;121:675–83. doi: 10.1161/CIRCULATIONAHA.109.902221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Schwarz ER, Pollick C, Dow J, Patterson M, Birnbaum Y, Kloner RA. A small animal model of nonischemic cardiomyopathy and its evaluation by transthoracic echocardiography. Cardiovasc Res. 1998;39:216–23. doi: 10.1016/s0008-6363(98)00009-1. [DOI] [PubMed] [Google Scholar]
  • 30.Lefrak EA, Pitha J, Rosenheim S, Gottlieb JA. A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer. 1973;32:302–14. doi: 10.1002/1097-0142(197308)32:2<302::aid-cncr2820320205>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 31.Adams MJ, Lipshultz SE. Pathophysiology of anthracycline- and radiation-associated cardiomyopathies: implications for screening and prevention. Pediatr Blood Cancer. 2005;44:600–6. doi: 10.1002/pbc.20352. [DOI] [PubMed] [Google Scholar]
  • 32.Yeh ET, Tong AT, Lenihan DJ, Yusuf SW, Swafford J, Champion C, et al. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation. 2004;109:3122–31. doi: 10.1161/01.CIR.0000133187.74800.B9. [DOI] [PubMed] [Google Scholar]
  • 33.Lipshultz SE, Adams MJ. Cardiotoxicity after childhood cancer: beginning with the end in mind. J Clin Oncol. 2010;28:1276–81. doi: 10.1200/JCO.2009.26.5751. doi: 10.00/JCO.2009.26.5751. Epub 2010 Feb 8. [DOI] [PubMed] [Google Scholar]
  • 34.Albini A, Pennesi G, Donatelli F, Cammarota R, De Flora S, Noonan DM. Cardiotoxicity of anticancer drugs: the need for cardio-oncology and cardio-oncological prevention. J Natl Cancer Inst. 2010;102:14–25. doi: 10.1093/jnci/djp440. doi: 10.1093/jnci/djp440. Epub 2009 Dec 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sawyer DB, Fukazawa R, Arstall MA, Kelly RA. Daunorubicin-induced apoptosis in rat cardiac myocytes is inhibited by dexrazoxane. Circ Res. 1999;84:257–65. doi: 10.1161/01.res.84.3.257. [DOI] [PubMed] [Google Scholar]
  • 36.Fisher PW, Salloum F, Das A, Hyder H, Kukreja RC. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation. 2005;111:1601–10. doi: 10.1161/01.CIR.0000160359.49478.C2. [DOI] [PubMed] [Google Scholar]
  • 37.Konorev EA, Vanamala S, Kalyanaraman B. Differences in doxorubicin-induced apoptotic signaling in adult and immature cardiomyocytes. Free Radic Biol Med. 2008;45:1723–8. doi: 10.1016/j.freeradbiomed.2008.09.006. doi: 10.016/j.freeradbiomed.2008.09.006. Epub Sep 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Czerski L, Nunez G. Apoptosome formation and caspase activation: is it different in the heart? J Mol Cell Cardiol. 2004;37:643–52. doi: 10.1016/j.yjmcc.2004.04.016. [DOI] [PubMed] [Google Scholar]
  • 39.Donovan D, Brown NJ, Bishop ET, Lewis CE. Comparison of three in vitro human 'angiogenesis' assays with capillaries formed in vivo. Angiogenesis. 2001;4:113–21. doi: 10.1023/a:1012218401036. [DOI] [PubMed] [Google Scholar]
  • 40.Gordon O, Gilon D, He Z, May D, Lazarus A, Oppenheim A, et al. Vascular endothelial growth factor-induced neovascularization rescues cardiac function but not adverse remodeling at advanced ischemic heart disease. Arterioscler Thromb Vasc Biol. 2012;32:1642–51. doi: 10.1161/ATVBAHA.112.248674. doi: 10.161/ATVBAHA.112.248674. Epub 2012 Apr 26. [DOI] [PubMed] [Google Scholar]
  • 41.Kairaitis LK, Wang Y, Gassmann M, Tay YC, Harris DC. HIF-1alpha expression follows microvascular loss in advanced murine adriamycin nephrosis. Am J Physiol Renal Physiol. 2005;288:F198–206. doi: 10.1152/ajprenal.00244.2003. Epub 2004 Sep 21. [DOI] [PubMed] [Google Scholar]
  • 42.Cheol Whee Park, Wha Chung Hyun, Wook Kim Hyung, Ju Lee Sang, Hee Lim Ji, Chae Chi-Bom, et al. Vascular Endothelial Growth Factor Inhibition by dRK6 Causes Endothelial Apoptosis, Fibrosis, and Inflammation in the Heart via the Akt/eNOS Axis in db/db Mice. Diabetes. 2009;58:2666–76. doi: 10.2337/db09-0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yoon YS, Uchida S, Masuo O, Cejna M, Park JS, Gwon HC, et al. Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy: restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor. Circulation. 2005;111:2073–85. doi: 10.1161/01.CIR.0000162472.52990.36. [DOI] [PubMed] [Google Scholar]
  • 44.Izumiya Y, Shiojima I, Sato K, Sawyer DB, Colucci WS, Walsh K. Vascular endothelial growth factor blockade promotes the transition from compensatory cardiac hypertrophy to failure in response to pressure overload. Hypertension. 2006;47:887–93. doi: 10.1161/01.HYP.0000215207.54689.31. Epub 2006 Mar 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shiojima I, Sato K, Izumiya Y, Schiekofer S, Ito M, Liao R, et al. Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. J Clin Invest. 2005;115:2108–18. doi: 10.1172/JCI24682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sano M, Minamino T, Toko H, Miyauchi H, Orimo M, Qin Y, et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature. 2007;446:444–8. doi: 10.1038/nature05602. Epub 2007 Mar 4. [DOI] [PubMed] [Google Scholar]
  • 47.De Angelis A, Piegari E, Cappetta D, Marino L, Filippelli A, Berrino L, et al. Anthracycline cardiomyopathy is mediated by depletion of the cardiac stem cell pool and is rescued by restoration of progenitor cell function. Circulation. 2010;121:276–92. doi: 10.1161/CIRCULATIONAHA.109.895771. doi: 10.1161/CIRCULATIONAHA.109.895771. Epub 2009 Dec 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Amoh Y, Li L, Katsuoka K, Hoffman RM. Chemotherapy targets the hair-follicle vascular network but not the stem cells. J Invest Dermatol. 2007;127:11–5. doi: 10.1038/sj.jid.5700486. Epub 2006 Jul 13. [DOI] [PubMed] [Google Scholar]
  • 49.Lorenzo E, Ruiz-Ruiz C, Quesada AJ, Hernandez G, Rodriguez A, Lopez-Rivas A, et al. Doxorubicin induces apoptosis and CD95 gene expression in human primary endothelial cells through a p53-dependent mechanism. J Biol Chem. 2002;277:10883–92. doi: 10.1074/jbc.M107442200. Epub 2002 Jan 4. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang L, Yu D, Hicklin DJ, Hannay JA, Ellis LM, Pollock RE. Combined anti-fetal liver kinase 1 monoclonal antibody and continuous low-dose doxorubicin inhibits angiogenesis and growth of human soft tissue sarcoma xenografts by induction of endothelial cell apoptosis. Cancer Res. 2002;62:2034–42. [PubMed] [Google Scholar]
  • 51.Kotamraju S, Konorev EA, Joseph J, Kalyanaraman B. Doxorubicin-induced apoptosis in endothelial cells and cardiomyocytes is ameliorated by nitrone spin traps and ebselen. Role of reactive oxygen and nitrogen species. J Biol Chem. 2000;275:33585–92. doi: 10.1074/jbc.M003890200. [DOI] [PubMed] [Google Scholar]
  • 52.Cleeland CS, Allen JD, Roberts SA, Brell JM, Giralt SA, Khakoo AY, et al. Reducing the toxicity of cancer therapy: recognizing needs, taking action. Nat Rev Clin Oncol. 2012;9:471–8. doi: 10.1038/nrclinonc.2012.99. doi: 10.1038/nrclinonc.2012.99. [DOI] [PubMed] [Google Scholar]
  • 53.Sheppard RJ, Berger J, Sebag IA. Cardiotoxicity of cancer therapeutics: current issues in screening, prevention, and therapy. Front Pharmacol. 2013;4:19. doi: 10.3389/fphar.2013.00019. 10.3389/fphar.2013.00019. Epub 2013 Mar 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mulrooney DA, Ness KK, Huang S, Solovey A, Hebbel RP, Neaton JD, et al. Pilot study of vascular health in survivors of osteosarcoma. Pediatr Blood Cancer. 2013;60:1703–8. doi: 10.1002/pbc.24610. doi: 10.002/pbc.24610. Epub 2013 May 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mulrooney DA, Ness KK, Solovey A, Hebbel RP, Neaton JD, Peterson BA, et al. Pilot study of vascular health in survivors of Hodgkin lymphoma. Pediatr Blood Cancer. 2012;59:285–9. doi: 10.1002/pbc.24082. doi: 10.1002/pbc.24082. Epub 2012 Mar 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Harris NL, Brenner DE, Anthony LB, Collins JC, Halter S, Hande KR. The influence of ranitidine on the pharmacokinetics and toxicity of doxorubicin in rabbits. Cancer Chemother Pharmacol. 1988;21:323–8. doi: 10.1007/BF00264199. [DOI] [PubMed] [Google Scholar]
  • 57.Konorev EA, Zhang H, Joseph J, Kennedy MC, Kalyanaraman B. Bicarbonate exacerbates oxidative injury induced by antitumor antibiotic doxorubicin in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2000;279:H2424–30. doi: 10.1152/ajpheart.2000.279.5.H2424. [DOI] [PubMed] [Google Scholar]
  • 58.Sarvazyan N. Visualization of doxorubicin-induced oxidative stress in isolated cardiac myocytes. Am J Physiol. 1996;271:H2079–85. doi: 10.1152/ajpheart.1996.271.5.H2079. [DOI] [PubMed] [Google Scholar]
  • 59.van Asperen J, van Tellingen O, Tijssen F, Schinkel AH, Beijnen JH. Increased accumulation of doxorubicin and doxorubicinol in cardiac tissue of mice lacking mdr1a P-glycoprotein. Br J Cancer. 1999;79:108–13. doi: 10.1038/sj.bjc.6690019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rahman A, Carmichael D, Harris M, Roh JK. Comparative pharmacokinetics of free doxorubicin and doxorubicin entrapped in cardiolipin liposomes. Cancer Res. 1986;46:2295–9. [PubMed] [Google Scholar]
  • 61.Lee YT, Chan KK, Harris PA. Tissue disposition of doxorubicin in experimental animals. Med Pediatr Oncol. 1982;10:259–67. doi: 10.1002/mpo.2950100306. [DOI] [PubMed] [Google Scholar]
  • 62.Duyndam MC, van Berkel MP, Dorsman JC, Rockx DA, Pinedo HM, Boven E. Cisplatin and doxorubicin repress Vascular Endothelial Growth Factor expression and differentially down-regulate Hypoxia-inducible Factor I activity in human ovarian cancer cells. Biochem Pharmacol. 2007;74:191–201. doi: 10.1016/j.bcp.2007.04.003. Epub 2007 Apr 6. [DOI] [PubMed] [Google Scholar]
  • 63.Lee K, Qian DZ, Rey S, Wei H, Liu JO, Semenza GL. Anthracycline chemotherapy inhibits HIF-1 transcriptional activity and tumor-induced mobilization of circulating angiogenic cells. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:2353–8. doi: 10.1073/pnas.0812801106. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 64.Chiusa M, Hool SL, Truetsch P, Djafarzadeh S, Jakob SM, Seifriz F, et al. Cancer therapy modulates VEGF signaling and viability in adult rat cardiac microvascular endothelial cells and cardiomyocytes. J Mol Cell Cardiol. 2012;52:1164–75. doi: 10.1016/j.yjmcc.2012.01.022. doi: 10.016/j.yjmcc.2012.01.022. Epub Feb 3. [DOI] [PubMed] [Google Scholar]
  • 65.Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S, Kalyanaraman B. Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J Biol Chem. 2004;279:25535–43. doi: 10.1074/jbc.M400944200. Epub 2004 Mar 30. [DOI] [PubMed] [Google Scholar]
  • 66.Frederick JP, Liberati NT, Waddell DS, Shi Y, Wang XF. Transforming growth factor beta-mediated transcriptional repression of c-myc is dependent on direct binding of Smad3 to a novel repressive Smad binding element. Mol Cell Biol. 2004;24:2546–59. doi: 10.1128/MCB.24.6.2546-2559.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Siegel PM, Massague J. Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer. 2003;3:807–21. doi: 10.1038/nrc1208. [DOI] [PubMed] [Google Scholar]
  • 68.Inman GJ, Nicolas FJ, Callahan JF, Harling JD, Gaster LM, Reith AD, et al. SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol Pharmacol. 2002;62:65–74. doi: 10.1124/mol.62.1.65. [DOI] [PubMed] [Google Scholar]
  • 69.Liu Z, Kobayashi K, van Dinther M, van Heiningen SH, Valdimarsdottir G, van Laar T, et al. VEGF and inhibitors of TGFbeta type-I receptor kinase synergistically promote blood-vessel formation by inducing alpha5-integrin expression. J Cell Sci. 2009;122:3294–302. doi: 10.1242/jcs.048942. doi: 10.1242/jcs.048942. Epub 2009 Aug 25. [DOI] [PubMed] [Google Scholar]
  • 70.Imaizumi N, Monnier Y, Hegi M, Mirimanoff RO, Ruegg C. Radiotherapy suppresses angiogenesis in mice through TGF-betaRI/ALK5-dependent inhibition of endothelial cell sprouting. PloS one. 2010;5:e11084. doi: 10.1371/journal.pone.0011084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Xavier S, Vasko R, Matsumoto K, Zullo JA, Chen R, Maizel J, et al. Curtailing Endothelial TGF-beta Signaling Is Sufficient to Reduce Endothelial-Mesenchymal Transition and Fibrosis in CKD. J Am Soc Nephrol. 2014;22:2013101137. doi: 10.1681/ASN.2013101137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.James D, Nam HS, Seandel M, Nolan D, Janovitz T, Tomishima M, et al. Expansion and maintenance of human embryonic stem cell-derived endothelial cells by TGFbeta inhibition is Id1 dependent. Nat Biotechnol. 2010;28:161–6. doi: 10.1038/nbt.1605. doi: 10.1038/nbt.605. Epub 2010 Jan 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Watabe T, Nishihara A, Mishima K, Yamashita J, Shimizu K, Miyazawa K, et al. TGF-beta receptor kinase inhibitor enhances growth and integrity of embryonic stem cell-derived endothelial cells. J Cell Biol. 2003;163:1303–11. doi: 10.1083/jcb.200305147. Epub 2003 Dec 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ifkovits JL, Addis RC, Epstein JA, Gearhart JD. Inhibition of TGFbeta signaling increases direct conversion of fibroblasts to induced cardiomyocytes. PLoS One. 2014;9:e89678. doi: 10.1371/journal.pone.0089678. doi: 10.1371/journal.pone.0089678. eCollection 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Suzuki A, Nagai T, Nishimatsu S, Sugino H, Eto Y, Shibai H, et al. Autoinduction of activin genes in early Xenopus embryos. A carboxyl-terminal truncated version of the activin receptor mediates activin signals in early Xenopus embryos. Biochem J. 1994;298:275–80. doi: 10.1042/bj2980275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kim SJ, Angel P, Lafyatis R, Hattori K, Kim KY, Sporn MB, et al. Autoinduction of transforming growth factor beta 1 is mediated by the AP-1 complex. Mol Cell Biol. 1990;10:1492–7. doi: 10.1128/mcb.10.4.1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yue J, Mulder KM. Requirement of Ras/MAPK pathway activation by transforming growth factor beta for transforming growth factor beta 1 production in a Smad-dependent pathway. J Biol Chem. 2000;275:30765–73. doi: 10.1074/jbc.M000039200. [DOI] [PubMed] [Google Scholar]
  • 78.Piek E, Ju WJ, Heyer J, Escalante-Alcalde D, Stewart CL, Weinstein M, et al. Functional characterization of transforming growth factor beta signaling in Smad2- and Smad3-deficient fibroblasts. J Biol Chem. 2001;276:19945–53. doi: 10.1074/jbc.M102382200. Epub 2001 Mar 21. [DOI] [PubMed] [Google Scholar]
  • 79.Strutz F, Zeisberg M, Renziehausen A, Raschke B, Becker V, van Kooten C, et al. TGF-beta 1 induces proliferation in human renal fibroblasts via induction of basic fibroblast growth factor (FGF-2). Kidney Int. 2001;59:579–92. doi: 10.1046/j.1523-1755.2001.059002579.x. [DOI] [PubMed] [Google Scholar]
  • 80.Clark RA, McCoy GA, Folkvord JM, McPherson JM. TGF-beta 1 stimulates cultured human fibroblasts to proliferate and produce tissue-like fibroplasia: a fibronectin matrix-dependent event. J Cell Physiol. 1997;170:69–80. doi: 10.1002/(SICI)1097-4652(199701)170:1<69::AID-JCP8>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
  • 81.Myeroff LL, Parsons R, Kim SJ, Hedrick L, Cho KR, Orth K, et al. A transforming growth factor beta receptor type II gene mutation common in colon and gastric but rare in endometrial cancers with microsatellite instability. Cancer Res. 1995;55:5545–7. [PubMed] [Google Scholar]
  • 82.Furuta K, Misao S, Takahashi K, Tagaya T, Fukuzawa Y, Ishikawa T, et al. Gene mutation of transforming growth factor beta1 type II receptor in hepatocellular carcinoma. Int J Cancer. 1999;81:851–3. doi: 10.1002/(sici)1097-0215(19990611)81:6<851::aid-ijc2>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
  • 83.Giannelli G, Villa E, Lahn M. Transforming growth factor-beta as a therapeutic target in hepatocellular carcinoma. Cancer Res. 2014;74:1890–4. doi: 10.1158/0008-5472.CAN-14-0243. doi: 10.158/0008-5472.CAN-14-0243. Epub 2014 Mar 17. [DOI] [PubMed] [Google Scholar]
  • 84.Tanaka K, Miyata H, Sugimura K, Fukuda S, Kanemura T, Yamashita K, et al. miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer- associated fibroblasts. Carcinogenesis. 2015;36:894–903. doi: 10.1093/carcin/bgv067. doi: 10.1093/carcin/bgv067. Epub 2015 May 30. [DOI] [PubMed] [Google Scholar]
  • 85.Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL. p38 mitogen-activated protein kinase is required for TGFbeta-mediated fibroblastic transdifferentiation and cell migration. J Cell Sci. 2002;115:3193–206. doi: 10.1242/jcs.115.15.3193. [DOI] [PubMed] [Google Scholar]
  • 86.Drabsch Y, He S, Zhang L, Snaar-Jagalska BE, ten Dijke P. Transforming growth factor-beta signalling controls human breast cancer metastasis in a zebrafish xenograft model. Breast Cancer Res. 2013;15:R106. doi: 10.1186/bcr3573. doi: 10.1186/bcr3573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hjelmeland MD, Hjelmeland AB, Sathornsumetee S, Reese ED, Herbstreith MH, Laping NJ, et al. SB-431542, a small molecule transforming growth factor-beta-receptor antagonist, inhibits human glioma cell line proliferation and motility. Mol Cancer Ther. 2004;3:737–45. [PubMed] [Google Scholar]
  • 88.Li J, Campanale NV, Liang RJ, Deane JA, Bertram JF, Ricardo SD. Inhibition of p38 mitogen-activated protein kinase and transforming growth factor-beta1/Smad signaling pathways modulates the development of fibrosis in adriamycin-induced nephropathy. Am J Pathol. 2006;169:1527–40. doi: 10.2353/ajpath.2006.060169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Li J, Deane JA, Campanale NV, Bertram JF, Ricardo SD. Blockade of p38 mitogen-activated protein kinase and TGF-beta1/Smad signaling pathways rescues bone marrow-derived peritubular capillary endothelial cells in adriamycin-induced nephrosis. J Am Soc Nephrol. 2006;17:2799–811. doi: 10.1681/ASN.2006020130. Epub 006 Sep 7. [DOI] [PubMed] [Google Scholar]
  • 90.Tsagalou EP, Anastasiou-Nana M, Agapitos E, Gika A, Drakos SG, Terrovitis JV, et al. Depressed coronary flow reserve is associated with decreased myocardial capillary density in patients with heart failure due to idiopathic dilated cardiomyopathy. Journal of the American College of Cardiology. 2008;52:1391–8. doi: 10.1016/j.jacc.2008.05.064. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

PCR array
Suppl. Fig. 1
Suppl. Fig. 2

RESOURCES