Abstract
Abnormally elevated bone morphogenetic protein 4 (BMP4) expression and mediated signaling play a critical role in the pathogenesis of chronic hypoxia-induced pulmonary hypertension (CHPH). In this study, we investigated the expression level and functional significance of four reported naturally occurring BMP4 antagonists, noggin, follistatin, gremlin1, and matrix gla protein (MGP), in the lung and distal pulmonary arterial smooth muscle cell (PASMC). A 21-day chronic hypoxic (10% O2) exposure rat model was utilized, which has been previously shown to successfully establish experimental CHPH. Among the four antagonists, noggin, but not the other three, was selectively downregulated by hypoxic exposure in both the lung tissue and PASMC, in correlation with markedly elevated BMP4 expression, suggesting that the loss of noggin might account for the hypoxia-triggered BMP4 signaling transduction. Then, by using treatment of extrogenous recombinant noggin protein, we further found that noggin significantly normalized 1) BMP4-induced phosphorylation of cellular p38 and ERK1/2; 2) BMP4-induced phosphorylation of cellular JAK2 and STAT3; 3) hypoxia-induced PASMC proliferation; 4) hypoxia-induced store-operated calcium entry (SOCE), and 5) hypoxia-increased expression of transient receptor potential cation channels (TRPC1 and TRPC6) in PASMC. In combination, these data strongly indicated that the hypoxia-suppressed noggin accounts, at least partially, for hypoxia-induced excessive PASMC proliferation, while restoration of noggin may be an effective way to inhibit cell proliferation by suppressing SOCE and TRPC expression.
Keywords: noggin, TRPC, store-operated calcium entry, pulmonary hypertension
pulmonary hypertension (PH) is a disease characterized by a list of functional and structural changes in the pulmonary vasculature that lead to enhanced distal pulmonary arterial (PA) contraction and remodeling, eventually causing heart failure. During the disease process, remodeling of small vessels in the lung, due to abnormal proliferation and migration of vascular smooth muscle cells and endothelium cells, is well studied and accepted (10).
Bone morphogenetic proteins (BMPs) belong to a subgroup of the transforming growth factor-β (TGF-β) superfamily, which are a group of growth factors originally discovered by their ability to induce the formation of bone and cartilage. Recently, evidence strongly indicated that dysregulated BMP signaling is involved in the pathogenesis of PH (9, 11). BMP4, a member of BMP ligands, has been found selectively upregulated by chronic hypoxia in the lungs and plays an important role during the development of chronic hypoxia-induced pulmonary hypertension (CHPH) by regulating the proliferation and migration of pulmonary arterial smooth muscle cells (PASMC) (11, 13, 20). In mechanisms, BMP4-mediated signaling transduction is mainly regulated by two groups of molecules, the typical receptors and the extracellular soluble antagonists (3). BMP4 transduces signals by binding to type II serine-threonine kinase receptors (44), which then causes recruitment and phosphorylation of type I receptors, leading to activation of a number of cellular kinases (18).
The group of BMP antagonists belongs to naturally secreted endogenous proteins that can block the BMP ligand-receptor interaction to inhibit the BMP signaling transduction. Increasing evidence demonstrated that dysfunction of these proteins leads to excessive BMP activity and signaling transduction, which is present and may account for the development of numerous diseases. In detail, gremlin1 specifically binds to BMP2, 4, and 7 and inhibits their actions on the downstream signaling (14, 23). Evidence suggests that gremlin1 is upregulated in lungs isolated from 2-day alveolar hypoxia-exposed mice (6). The increased gremlin1 expression mostly localizes in the pulmonary endothelium, but not smooth muscle (2). Deletion of gremlin1 increases cell proliferation and migration responses in mouse embryonic fibroblasts (7). Noggin, follistatin, and matrix gla protein (MGP) are all demonstrated as BMP4 antagonists, which are found to coexpress with BMP4 at sites of oscillatory shear stress in the systemic vasculature (4). Noggin has long been known as a classic BMP antagonist with a high-affinity binding to BMP4 (25, 26, 49). Traditionally defined as an antagonist of activin protein, follistatin could also interact with BMPs (including BMP4, 5, 6, 7, and 15), though in a lower-affinity range (5, 12, 32, 35). MGP is an extracellular matrix component expressing high abundance in vascular smooth muscle cells. MGP has been defined as contributing to the development of vasculature by using MGP-deficient mice (45). MGP inhibits or activates BMPs (BMP2 and BMP4) in a concentration-dependent manner (46, 48). So far, many groups have discussed the effects of BMPs antagonists on interfering BMP signaling and participating in the development of different diseases. However, the full action of these members in CHPH remains largely unknown. We previously demonstrated that animals exposed to chronic hypoxia (CH, 10% O2) for 21 days were used as an animal model for mimicking CHPH (21, 40). Thus, in this study, we aimed to figure out the changes in these antagonists upon hypoxic stresses and determine their roles in the process of hypoxia-induced proliferation in PASMC.1
MATERIALS AND METHODS
Chronic hypoxic exposure of rats and primary culture of PASMC.
All animal experimental procedures were approved by the Institutional Animal Care and Use Committee of The Johns Hopkins University School of Medicine. The surgical procedure was performed under anesthesia with pentobarbital sodium (65 mg/kg ip), and all efforts were made to minimize animal suffering. Adult male Wistar rats (150–250 g) were purchased from Harlan (Frederick, MD). Rats were randomly divided into two groups and exposed to normoxia or chronic hypoxia (10% O2) for 21 days as previously described (34, 40). The hypoxic chamber was continuously flushed with a mixture of room air and N2 to maintain 10 ± 0.5% O2 and CO2 <0.5%. Chamber O2 concentration was continuously monitored using a PRO-OX unit (RCI Hudson, Anaheim, CA). Rats were exposed to room air for 10 min every day for administering food and water, or change of cage. By the end of the exposure, rats from all groups were anesthetized with pentobarbital sodium (65 mg/kg ip) and euthanized for harvesting lung tissues. The blood in the vessel was gently completely washed out with PBS to eliminate the interference of the circulation. The primary culture of distal PASMC was based on an enzymatic digestion method, as previously described (39). Basically, distal (>4th generation) intralobar distal PAs were dissected from the lungs. Adventitia was removed from the isolated PAs, and endothelium was denuded by opening the vessel longitudinally and rubbing the luminal surface with a cotton swab.
Real-time quantitative polymerase chain reaction.
Total RNA was extracted using TRIzol method for lung tissues, as previously described (21). DNA contamination in RNA preparations was removed by on-column DNase digestion using RNeasy column and RNase-free DNase (Qiagen, Valencia, CA). Reverse transcription was performed using iScript cDNA synthesis kit (Bio-Rad, Hercules, CA) with reaction mixture containing 1 μg total RNA in a 20 μl volume. cDNA were amplified by real-time quantitative polymerase chain reaction (RT-qPCR) using QuantiTect SYBR Green PCR Master Mix (Qiagen) in an iCyclerIQ detection system (Bio-Rad). The protocol consisted of initial enzyme activation at 95°C for 3 min, followed by 40 cycles at 95°C for 5 s and at 60°C for 15 s (22). Primer sequences were designed using Primer3 software and are listed in Table 1. Relative concentration of each transcript was calculated using the Pfaffl method (33). Efficiency for each gene was determined from 5-point serial dilutions of an unknown cDNA sample. The expression of BMP antagonists was normalized to cyclophilin B as internal control.
Table 1.
Primers for real time-qPCR
| Gene | Accession No. | Source | Primer Sequence (left/right) | Product Size, bp |
|---|---|---|---|---|
| Follistatin | NM_012561.1 | Rat | 5′-AACCTACCGCAACGAATGTG-3′ | 138 |
| 5′-TGATCCACCACACAAGTGGA-3′ | ||||
| Noggin | NM_012990.1 | Rat | 5′-CCTGGCTTTCTGGTTCATGT-3′ | 123 |
| 5′-GCCGGGTAACTTTTGACGTA-3′ | ||||
| Gremlin1 | NM_019282.3 | Rat | 5′-GACAAGGCTCAGCACAATGA-3′ | 118 |
| 5′-ACTCAAGCACCTCCTCTCCA-3′ | ||||
| MGP | NM_012862.1 | Rat | 5′-TGAATCTCACGAAAGCATGG-3′ | 91 |
| 5′-CCATCTCTGCTGAGGGGATA-3′ | ||||
| Cyclophilin B | NM_022536.1 | Rat | 5′-CAAGACCTCCTGGCTAGACG-3′ | 81 |
| 5′-TTCTCCACCTTCCGTACCAC-3′ |
Western blotting.
Lung tissue samples were sonicated or homogenized in T-PER sample buffer (Pierce, Rockford, IL) containing protease inhibitor cocktail. Total protein concentration in the homogenates was determined by bicinchoninic acid protein assay (Pierce) using bovine serum albumin as a standard. Homogenates were denatured by adding dithiothreitol to 150 mM and heating at 95°C for 3 min. Homogenate proteins were resolved by 10% SDS-PAGE calibrated with Precision Plus protein molecular weight markers (Bio-Rad). Separated proteins were transferred to 0.45 μM polyvinylidene difluoride membranes (Bio-Rad). Membranes were blocked with 5% nonfat dry milk in Tris-buffered saline containing 0.2% Tween 20, and blotted with affinity-purified polyclonal antibodies specific for TRPC1 and TRPC6 (Sigma, St. Louis, MO), MGP (Proteintech, Chicago, IL), noggin (BD Biosciences, San Jose, CA), follistatin (Abcam, Cambridge, MA), gremlin1 (Abcam), p-p38, t-p38, p-ERK1/2, t-ERK1/2, p-JAK2, and p-STAT3 (Cell Signaling Technology, Beverly, MA) or monoclonal antibody against β-tubulin and β-actin (Sigma). The membranes were then washed for 10 min 3 times and incubated with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Kirkegaard and Perry Laboratories, Gaithersburg, MD) for 1 h. Bound antibodies were detected using an enhanced chemiluminescence system (ECL, GE Healthcare, Piscataway, NJ).
Measurement of store-operated calcium entry.
Following previously described procedures (39), cultured PASMC were seeded on coverslips and serum starved for 24 h in smooth muscle basal medium (SMBM) containing 0.3% FBS and 1-h loading with 7.5 μM fluorescent dye fura-2 AM (Molecular Probes, Eugene, OR) before calcium measurement experiments. Intracellular Ca2+ concentration was calculated by the ratio of fura-2 fluorescence emitted at 510 nm after excitation at 340 nm to that after excitation at 380 nm (F340/F380). Store-operated calcium entry (SOCE) was evaluated by extracellular calcium restoration method, in which intracellular sarcoplasmic reticulum calcium stores within PASMC were depleted by perfusion of 10 μM cyclopiazonic acid (CPA), in the presence of 1 mM EGTA and 5 μM voltage-dependent calcium channel (VDCC) blocker nifedipine to exclude the influence of VDCC. By restoring extracellular calcium we could measure SOCE through store-operated calcium channels (SOCC).
Measurement of PASMC proliferation.
The proliferation of PASMC was measured with the Cell Proliferation Biotrak ELISA Kit (GE Healthcare) based on a BrdU incorporation assay, as previously described (41). Briefly, PASMC were seeded in 96-well plates in SMBM at a density of 5 × 103 cells/well. After each different treatment, the cells were then labeled with BrdU for 24 h, fixed and blocked; antibody was probed, developed with TMB substrate, and stopped with sulfuric acid, following kit instructions. The optical density of wells was measured with a microplate reader (Bio-Rad) at 450 nm.
Materials and reagents.
Unless otherwise specified, all the reagents were obtained from Sigma-Aldrich. Fura-2 AM (Invitrogen) was prepared before the experiment as a 2.5 mM stock solution in 20% DMSO containing 20% pluronic F-127 (Invitrogen). Stock solutions of nifedipine and CPA were both made in DMSO at 30 mM. Human recombinant BMP4 protein was purchased from R&D and noggin recombinant protein was purchased from Invitrogen.
Statistical analysis.
All of the data are represented as means ± SE; n is number of experiments, which equals the number of animals providing PA or lung; number of dishes of cells for in vitro molecular biological experiments and intracellular calcium imaging experiments. Statistical analyses were performed using analysis of Students t-test and one-way ANOVA. Pairwise comparison of means was conducted with t-tests. For the groups containing multiple comparisons, one-way ANOVA was used for the statistical analysis. Differences were considered significant when P < 0.05.
RESULTS
Chronic hypoxic exposure (10% O2, 21-day) selectively downregulates noggin expression in rat lung tissues.
First, we focused on the mRNA and protein expression of the four BMP antagonists noggin, follistatin, gremlin1, and MGP in the lungs isolated from both normoxic and CH-exposed rats. We designed specific primers for all of these members for real-time qPCR amplification (Table 1). By using real-time qPCR, we found that all of the four BMP antagonists were expressed in rat lung tissue and among which, noggin was selectively downregulated for 41.9% by hypoxic exposure (Fig. 1A). Then, by using Western blotting, we found that noggin protein levels were also significantly downregulated 45.7% (Fig. 1, B and C) by CH exposure, in comparison with normoxic control. However, the other three antagonists remained unaltered by hypoxic stress in the lung.
Fig. 1.
Chronic hypoxia (10% O2, 21-day) downregulates noggin expression at both mRNA and protein levels in rat lung tissue. A: bar graph representing mRNA fold induction of noggin, follistatin, gremlin1, and matrix gla protein (MGP) in lungs isolated from 21-day chronic hypoxia-exposed rats normalized to levels from normoxic control rats. Cyclophilin B (CpB) served as housekeeping internal control. B and C: Western blots (B) and bar graph (C) showing protein expression levels of the four bone morphogenetic protein (BMP) antagonists in rat lung tissues under normoxic and hypoxic conditions. β-Actin served as housekeeping protein. S1, S2, and S3 mean the number of each individual sample in each group of treatment. The bar graph represents means ± SE; n = 6–12 in each group. *P < 0.05 vs. normoxic control.
Prolonged hypoxic exposure (4% O2, 60-h) selectively downregulates noggin expression in rat distal PASMC.
Since previous studies strongly indicated that the excessive proliferation and migration of distal PASMC act as two major events contributing to distal PA remodeling and thickening during PH pathogenesis, we further explored if expression of these antagonists can also be regulated by hypoxia in cultured PASMC. Primary cultured rat distal PASMC were serum starved and exposed to prolonged hypoxia (4% O2) for up to 60 h, the period of which has been proved to induce elevation of intracellular calcium concentration and cell proliferation (40). We used real-time PCR to measure mRNA transcription levels and Western blotting to measure protein levels of the four BMP antagonists in PASMC upon prolonged hypoxic exposure. The RT-qPCR data showed no obvious change in the mRNA levels of all these four antagonists under different time points of hypoxic exposure (Fig. 2, A–D). However, the protein level of noggin, but not the others, was selectively reduced at the 48- and 60-h hypoxic exposure time points, accompanied with upregulated BMP4 protein levels (Fig. 2, E and F). These results indicate the possibility that the hypoxia-suppressed noggin, acting in correlation with the hypoxia-induced BMP4, might account for the uncontrolled proliferation and elevated intracellular calcium concentration and SOCE in PASMC.
Fig. 2.
Prolonged hypoxia (4% O2) inhibits noggin protein expression in rat distal pulmonary arterial smooth muscle cells (PASMC). A–D: bar graphs representing mRNA fold induction of noggin, follistatin, gremlin1, and MGP in rat distal PASMC that underwent hypoxic exposure (H) for 6–60 h, as relevant to CpB. NOR, normoxia. E and F: Western blots (E) and line chart (F) showing protein expression levels of the four BMP antagonists and BMP4 in cultured rat distal PASMC under different time points of hypoxic exposure. The bar graph represents means ± SE; n = 3–6 in each group. *P < 0.05 vs. normoxic control (BMP4 line); #P <0.05 vs. normoxic control (noggin line).
Treatment of extrogenous recombinant noggin inhibits BMP4-induced phosphorylation of p38 and ERK1/2 in PASMC.
Our previous study indicated that among the substantial cellular signaling pathways that can be induced by BMP4, the p38 and ERK1/2 were considered to be essential for the induction of downstream SOCE processes and the expression of SOCC components TRPC1 and TRPC6 (18). Thus, we applied treatment of exogenous recombinant noggin protein and detected if noggin can block BMP4-induced p38 and ERK1/2 activation. Our results showed that 200 ng/ml of noggin treatment totally blocked BMP4-induced phosphorylation of p38 and ERK1/2 (Fig. 3, A and B).
Fig. 3.
Noggin blocks BMP4-induced p38 and ERK1/2 phosphorylation in PASMC. A: Western blots showing protein expression levels of p-p38, t-p38, p-ERK1/2, t-ERK1/2, and β-tubulin in cultured rat distal PASMC upon BMP4 (50 ng/ml) treatment for 15 min after pretreatment with noggin (50 and 200 ng/ml) for 30 min. B: bar graphs representing the phosphorylation rates of p-p38 (left) and p-ERK1/2 (right) as normalized to t-p38 and t-ERK1/2. The bar graph represents means ± SE; n = 3 in each group. *P < 0.05 vs. vehicle control; #P < 0.05 vs. BMP4 (50 ng/ml)-treated group.
Treatment of extrogenous recombinant noggin inhibits BMP4-induced phosphorylation of JAK2 and STAT3 in PASMC.
As we know, p38 and ERK are not the only kinases to initiate and mediate the cellular pro-proliferation subsequences. Actually, a variety of previous publications have reported that the JAK2/STAT3 signaling axis can mediate smooth muscle cell proliferation upon activation from upstream stimulation (1, 24, 36–38). Therefore, we found that BMP4 treatment (50 ng/ml) can activate the JAK2/STAT3 signaling cascade in a time-dependent manner (Fig. 4, A and B), then, a pretreatment with noggin (200 ng/ml) for 30 min significantly attenuated the BMP4-induced phosphorylation of JAK2/STAT3 (Fig. 4, C and D).
Fig. 4.
Noggin attenuates BMP4-induced JAK2 and STAT3 phosphorylation in PASMC. A and B: BMP4 time dependently induces JAK2 and STAT3 phosphorylation in PASMC. Western blots show protein expression levels of p-JAK2, p-STAT3, and β-tubulin in cultured rat distal PASMC upon BMP4 (50 ng/ml) treatment for 5–120 min (A). Bar graphs represent the phosphorylation rates of p-JAK2 (left) and p-STAT3 (right) as normalized to β-tubulin (B). C and D: pretreatment of noggin (200 ng/ml) for 30 min attenuates BMP4 (50 ng/ml, 30-min)-induced phosphorylation of JAK2/STAT3 in PASMC. Western blots show protein expression levels of p-JAK2, p-STAT3, and β-tubulin in cultured rat distal PASMC upon BMP4 and noggin treatments (C). Bar graphs represent the phosphorylation rates of p-JAK2 (left) and p-STAT3 (right) as normalized to β-tubulin (D). The bar graph represents means ± SE; n = 3–5 in each group. *P < 0.05 vs. control; #P < 0.05 vs. BMP4 (50 ng/ml)-treated group.
Treatment of extrogenous recombinant noggin inhibits hypoxia-elevated proliferation of PASMC.
Since the dosage of 200 ng/ml of noggin treatment totally blocks BMP4-induced p38 and ERK1/2 activation, we further determined the effect of 200 ng/ml noggin on the proliferation in PASMC under both normoxic and prolonged hypoxic conditions. Our results showed that first, hypoxia markedly increased the proliferation rate 41.6% compared with normoxic control. Then, 200 ng/ml noggin significantly normalized hypoxia-elevated proliferation to basal levels, without altering the proliferation under normoxia (Fig. 5).
Fig. 5.

Noggin normalized hypoxia-elevated proliferation in PASMC. The bar graph represents the fold induction of the PASMC proliferation rate in response to prolonged hypoxia (4% O2, 60-h) exposure with or without noggin (200 ng/ml) treatment, as normalized to normoxic control value. The bar graph represents means ± SE; n = 4 in each group. *P <0 .05 vs. normoxic control; #P < 0.05 vs. hypoxic control.
Noggin attenuates hypoxia-elevated SOCE in PASMC.
Furthermore, we also detected the role of noggin treatment on the SOCE and intracellular calcium regulation in PASMC, since hypoxia-enhanced SOCE is considered a key regulator to promote PASMC proliferation and PA remodeling (40). Our results indicated that prolonged hypoxia significantly enhanced SOCE from 295.9 nM to 372.7 nM, while noggin dramatically normalized hypoxia-enhanced SOCE to 275.7 nM, without markedly altering the SOCE under normoxia (Fig. 6).
Fig. 6.
Noggin attenuates hypoxia-elevated store-operated calcium entry (SOCE) in PASMC. A: line charts showing the dynamic traces of the intracellular calcium concentration ([Ca2+]i) in response to intracellular calcium store depletion by cyclopiazonic acid (CPA, 10 μM), in the presence of the voltage-dependent calcium channel (VDCC) blocker nifedipine (Nif; 5 μM) in calcium-free solution (0 Ca2+) and restoration of extracellular calcium concentration afterward. B: bar graph indicating the Δ changes before and after extracellular calcium restoration was calculated as SOCE. The bar graph represents means ± SE; n = 3 experiments in each group. Four groups were designed as normoxic control (Nor), normoxia+noggin (Nor+Nog, 200 ng/ml, 60 h), hypoxic control (Hyp), and hypoxia+noggin (Hyp+Nog, 200 ng/ml, 60 h). *P < 0.05 vs. normoxic control; #P < 0.05 vs. hypoxic control.
Noggin normalizes hypoxia-induced upregulation of TRPC1 and TRPC6 in PASMC.
Given the fact that noggin significantly normalized hypoxia-induced cell proliferation and SOCE in PASMC, we therefore investigated the role of noggin on the expression of the main SOCC components TRPC1 and TRPC6, which was previously reported to mediate hypoxia-enhanced SOCE in PASMC. As seen in Fig. 7, noggin treatment also attenuates the upregulated TRPC1 and TRPC6 protein expression under hypoxic condition, but not under normoxia.
Fig. 7.
Noggin normalized hypoxic upregulation of transient receptor potential cation channels TRPC1 and TRPC6 protein expression in PASMC. A: Western blots showing protein expression levels of TRPC1, TRPC6, and β-tubulin in cultured rat distal PASMC upon hypoxic exposure (4% O2, 60 h) and noggin (200 ng/ml) treatment. B: bar graphs representing the fold induction of TRPC1 (left) and TRPC6 (right) under different treatments, as normalized to normoxic control. β-Tubulin served as internal control. The bar graph represents means ± SE; n = 3 in each group. *P < 0.05 vs. normoxic control; #P < 0.05 vs. hypoxic control.
DISCUSSION
Previous evidence strongly indicated the essential contribution of increased BMP4 expression and elevated BMP4-mediated signaling transduction in CHPH pathogenesis. In this study, we investigated the expression and role of a group of four reported endogenous BMP4 antagonists in response to hypoxic stress in rat lung and distal PASMC. Our results indicated that among these four antagonists, noggin was selectively downregulated by hypoxic exposure in both lung and PASMC, in correlation with hypoxia-elevated BMP4 expression and activity. On the basis of the previous data, we hypothesized that hypoxia-suppressed noggin lacks the ability as an antagonist to inhibit the hypoxia-evoked BMP4 activity, leading to promoted BMP4 signaling transduction, which results in the transcription of numerous proliferative genes that triggers the cellular pro-proliferative subsequences. We further observed that extrogenous noggin treatment significantly normalized hypoxia-triggered proliferation, SOCE, and TRPC expression, suggesting that suppressed noggin contributes to the elevated BMP4 signaling and activity under hypoxia.
BMPs belong to the TGF-β superfamily, which are a group of factors originally found to induce ectopic bone formation when implanted subcutaneously (42). Then, considerable evidence demonstrated that BMPs also act as key multifunctional regulators in regulating cell proliferation, differentiation, and apoptosis in different tissues (27). BMPs exert their signals via binding with type II transmembrane serine/threonine kinase receptors, thus recruit with type 1 transmembrane serine/threonine kinase receptors, then activating the intracellular Smad-dependent and Smad-independent (e.g., ERK, JNK, and p38 MAP kinase pathways) signaling pathways in regulating substantial downstream gene expression (43). Basically, three type II receptors (BMPRII, ActRIIa, and ActRIIb) and three type I receptors (ALK2, ALK3, and ALK6) have already been indicated to be able to interfere with BMP ligands and transduce signaling (27, 28). Recently, a number of studies suggested that abnormal BMP signaling participates in the disease pathogenesis of PH. Many works have been done based on the finding of large chances of BMPRII mutation in idiopathic PAH and familial PAH patients (8, 17).
Besides the specific BMP receptors, there are also several extracellular modulators that can interfere with the BMPs ligand-receptor binding to block BMP signaling. Noggin, MGP, gremlin1, and follistatin belong to such members. These soluble antagonists dampen the BMP signaling either by competitively interacting with the BMP ligands and protecting them from binding with their specific receptors or by blocking the intracellular signal transduction without affecting ligand-receptor reaction (3, 15, 49). Previous publications addressed the different localization of these antagonists in the lung. Gremlin1 is identified in the endothelium, proximal airway epithelium, and alveolar epithelium (19, 29). Follistatin is widely expressed in bronchial epithelial cells, alveolar macrophages, and vascular smooth muscle cells (47). MGP is excessively and broadly expressed in the lung tissue (48). As it is well accepted that dysfunction of BMP4 signaling participates in and contributes to the excessive proliferation of PASMC in PH, we wondered whether the triggered BMP4 signaling is possibly, to some extent, due to the loss of expression and function of the antagonists.
Hence, we investigated the expression changes of these antagonists in response to hypoxic exposure in both the lung and PASMC. Our results showed that among these four antagonists, noggin was selectively downregulated by hypoxia in both the lung and PASMC. However, by applying the extrogenous noggin recombinant protein treatment, we further demonstrated that under hypoxia, the restoration of noggin effectively normalized hypoxia-induced proliferation, SOCE, and TRPC expression. It is important to compare the blockage efficiency between noggin treatment and direct BMP4 inhibition strategy (BMP4 knockdown). Actually, in one of our previous publications (20), we reported that knockdown of BMP4 by using specific siRNA effectively leads to dramatic decrease in the hypoxia-elevated TRPC-SOCE-[Ca2+]i signaling axis. Given the fact that the intracellular free calcium concentration acts as a major factor to enter the nucleus, facilitiates the transcription of a number of pro-proliferative genes and promote cell proliferation (16, 30, 31), we can conclude that in PASMC, BMP4 is responsible for the proliferation and intracellular calcium homeostasis, especially the SOCE process. Moreover, in this study, we further showed that noggin exerts similar inhibitory roles, in comparison with direct BMP4 knockdown, on the TRPC-SOCE-proliferation signaling axis. Therefore, our data suggest that a decrease in the naturally endogenous expressing BMP4 antagonist noggin is potentially a main reason accounting for the elevated BMP4 signaling pathway. The hypoxia-inhibited noggin accounts for, at least partly, the hypoxia-triggered BMP4 activity and signaling transduction. Due to the lack of noggin's protective role, evoked BMP4 signaling leads to upregulated TRPC-SOCE axis, which eventually results in excessive PASMC proliferation in PH development. Specific noggin-targeted restoration might act as a potential novel strategy to modulate BMP4 expression and activity, thus mediating an attenuation of hypoxia-elevated proliferation in PASMC. Moreover, we also found that besides the p38/ERK-TRPC-SOCE-[Ca2+]i signaling axis, BMP4 can also induce PASMC proliferation by inducing activation of JAK2/STAT3 signaling cascade. Our results strongly indicate that noggin can effectively attenuate BMP4-mediated proliferation by inhibiting both p38/ERK-TRPC-SOCE targeted calcium-dependent signaling axis and JAK2/STAT3 targeted calcium independent signaling axis.
In summary, this study presents initial evidence for the expression pattern of the four BMPs antagonists in response to hypoxic stress. We observed that the hypoxia-inhibited noggin level acts as a key element to result in excessive PASMC proliferation due to the uncontrolled elevated BMP4 signaling, as well as the downstream triggered p38/ERK-mediated calcium-dependent and JAK2/STAT3-mediated calcium-independent pro-proliferative signaling pathways, while strategies targeting noggin restoration might be a useful way to attenuate the hypoxia-elevated proliferation of PASMC. This study provides evidence of a novel potential target, noggin, which can potentially attenuate the hypoxia-elevated proliferation of PASMC and deserves further study and research.
GRANTS
This work was supported by National Institutes of Health (National Heart, Lung, and Blood Institute Grant R01 HL-093020), National Natural Science Foundation of China (81173112, 81470246, 81170052, 81220108001), Guangzhou Department of Education Yangcheng Scholarship (12A001S), Guangzhou Department of Natural Science (2014Y2-00167) and Guangdong Province Universities and Colleges Pearl River Scholar Funded Scheme (2014, W. Lu), and China Scholarship Council (201208440091 for K. Yang).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
AUTHOR CONTRIBUTIONS
K.Y., J.J., J.Z., H.J., and L.X. performed experiments; K.Y., W.L., and J.W. interpreted results of experiments; K.Y., J.J., and J.W. prepared figures; K.Y. drafted manuscript; J.J., J.Z., W.L., and J.W. analyzed data; M.Z., S.W., W.L., and J.W. edited and revised manuscript; W.L. and J.W. conception and design of research; W.L. and J.W. approved final version of manuscript.
Footnotes
This article is the topic of an Editorial Focus by Olivier Boucherat and Sébastien Bonnet (1a).
REFERENCES
- 1.Banes AK, Shaw SM, Tawfik A, Patel BP, Ogbi S, Fulton D, Marrero MB. Activation of the JAK/STAT pathway in vascular smooth muscle by serotonin. Am J Physiol Cell Physiol 288: C805–C812, 2005. [DOI] [PubMed] [Google Scholar]
- 1a.Boucherat O, Bonnet S. NOGGIN: a new therapeutic target for PH? Focus on “Noggin inhibits hypoxia-induced proliferation by targeting store-operated calcium entry and transient receptor potential cation channels.” Am J Physiol Cell Physiol. doi: 10.1152/ajpcell.00088.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cahill E, Costello CM, Rowan SC, Harkin S, Howell K, Leonard MO, Southwood M, Cummins EP, Fitzpatrick SF, Taylor CT, Morrell NW, Martin F, McLoughlin P. Gremlin plays a key role in the pathogenesis of pulmonary hypertension. Circulation 125: 920–930, 2012. [DOI] [PubMed] [Google Scholar]
- 3.Canalis E, Economides AN, Gazzerro E. Bone morphogenetic proteins, their antagonists, and the skeleton. Endocr Rev 24: 218–235, 2003. [DOI] [PubMed] [Google Scholar]
- 4.Chang K, Weiss D, Suo J, Vega JD, Giddens D, Taylor WR, Jo H. Bone morphogenic protein antagonists are coexpressed with bone morphogenic protein 4 in endothelial cells exposed to unstable flow in vitro in mouse aortas and in human coronary arteries: role of bone morphogenic protein antagonists in inflammation and atherosclerosis. Circulation 116: 1258–1266, 2007. [DOI] [PubMed] [Google Scholar]
- 5.Chen YG, Lui HM, Lin SL, Lee JM, Ying SY. Regulation of cell proliferation, apoptosis, and carcinogenesis by activin. Exp Biol Med (Maywood) 227: 75–87, 2002. [DOI] [PubMed] [Google Scholar]
- 6.Costello CM, Howell K, Cahill E, McBryan J, Konigshoff M, Eickelberg O, Gaine S, Martin F, McLoughlin P. Lung-selective gene responses to alveolar hypoxia: potential role for the bone morphogenetic antagonist gremlin in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 295: L272–L284, 2008. [DOI] [PubMed] [Google Scholar]
- 7.Curran SP, Hickey FB, Watson A, Godson C, Brazil DP. Deletion of Gremlin1 increases cell proliferation and migration responses in mouse embryonic fibroblasts. Cell Signal 24: 889–898, 2012. [DOI] [PubMed] [Google Scholar]
- 8.Deng Z, Morse JH, Slager SL, Cuervo N, Moore KJ, Venetos G, Kalachikov S, Cayanis E, Fischer SG, Barst RJ, Hodge SE, Knowles JA. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet 67: 737–744, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Eickelberg O, Morty RE. Transforming growth factor beta/bone morphogenic protein signaling in pulmonary arterial hypertension: remodeling revisited. Trends Cardiovasc Med 17: 263–269, 2007. [DOI] [PubMed] [Google Scholar]
- 10.Farber HW, Loscalzo J. Pulmonary arterial hypertension. N Engl J Med 351: 1655–1665, 2004. [DOI] [PubMed] [Google Scholar]
- 11.Frank DB, Abtahi A, Yamaguchi DJ, Manning S, Shyr Y, Pozzi A, Baldwin HS, Johnson JE, de Caestecker MP. Bone morphogenetic protein 4 promotes pulmonary vascular remodeling in hypoxic pulmonary hypertension. Circ Res 97: 496–504, 2005. [DOI] [PubMed] [Google Scholar]
- 12.Glister C, Kemp CF, Knight PG. Bone morphogenetic protein (BMP) ligands and receptors in bovine ovarian follicle cells: actions of BMP-4, -6 and -7 on granulosa cells and differential modulation of Smad-1 phosphorylation by follistatin. Reproduction 127: 239–254, 2004. [DOI] [PubMed] [Google Scholar]
- 13.Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM, Guignabert C, Bekker JM, Schellong S, Urashima T, Wang L, Morrell NW, Rabinovitch M. An antiproliferative BMP-2/PPARgamma/apoE axis in human and murine SMCs and its role in pulmonary hypertension. J Clin Invest 118: 1846–1857, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hsu DR, Economides AN, Wang X, Eimon PM, Harland RM. The Xenopus dorsalizing factor Gremlin identifies a novel family of secreted proteins that antagonize BMP activities. Mol Cell 1: 673–683, 1998. [DOI] [PubMed] [Google Scholar]
- 15.Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume T, Shimasaki S, Sugino H, Ueno N. Direct binding of follistatin to a complex of bone-morphogenetic protein and its receptor inhibits ventral and epidermal cell fates in early Xenopus embryo. Proc Natl Acad Sci USA 95: 9337–9342, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jernigan NL, Resta TC. Calcium homeostasis and sensitization in pulmonary arterial smooth muscle. Microcirculation 21: 259–271, 2014. [DOI] [PubMed] [Google Scholar]
- 17.Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA 3rd, Loyd JE, Nichols WC, Trembath RC. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet 26: 81–84, 2000. [DOI] [PubMed] [Google Scholar]
- 18.Li X, Lu W, Fu X, Zhang Y, Yang K, Zhong N, Ran P, Wang J. BMP4 increases canonical transient receptor potential protein expression by activating p38 MAPK and ERK1/2 signaling pathways in pulmonary arterial smooth muscle cells. Am J Respir Cell Mol Biol 49: 212–220, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lu MM, Yang H, Zhang L, Shu W, Blair DG, Morrisey EE. The bone morphogenic protein antagonist gremlin regulates proximal-distal patterning of the lung. Dev Dyn 222: 667–680, 2001. [DOI] [PubMed] [Google Scholar]
- 20.Lu W, Ran P, Zhang D, Lai N, Zhong N, Wang J. Bone morphogenetic protein 4 enhances canonical transient receptor potential expression, store-operated Ca2+ entry, and basal [Ca2+]i in rat distal pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 299: C1370–C1378, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lu W, Ran P, Zhang D, Peng G, Li B, Zhong N, Wang J. Sildenafil inhibits chronically hypoxic upregulation of canonical transient receptor potential expression in rat pulmonary arterial smooth muscle. Am J Physiol Cell Physiol 298: C114–C123, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lu W, Wang J, Shimoda LA, Sylvester JT. Differences in STIM1 and TRPC expression in proximal and distal pulmonary arterial smooth muscle are associated with differences in Ca2+ responses to hypoxia. Am J Physiol Lung Cell Mol Physiol 295: L104–L113, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Maciel TT, Melo RS, Schor N, Campos AH. Gremlin promotes vascular smooth muscle cell proliferation and migration. J Mol Cell Cardiol 44: 370–379, 2008. [DOI] [PubMed] [Google Scholar]
- 24.Madamanchi NR, Li S, Patterson C, Runge MS. Thrombin regulates vascular smooth muscle cell growth and heat shock proteins via the JAK-STAT pathway. J Biol Chem 276: 18915–18924, 2001. [DOI] [PubMed] [Google Scholar]
- 25.McMahon JA, Takada S, Zimmerman LB, Fan CM, Harland RM, McMahon AP. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev 12: 1438–1452, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mehler MF, Mabie PC, Zhang D, Kessler JA. Bone morphogenetic proteins in the nervous system. Trends Neurosci 20: 309–317, 1997. [DOI] [PubMed] [Google Scholar]
- 27.Miyazono K, Kamiya Y, Morikawa M. Bone morphogenetic protein receptors and signal transduction. J Biochem 147: 35–51, 2010. [DOI] [PubMed] [Google Scholar]
- 28.Moustakas A, Heldin CH. The regulation of TGFbeta signal transduction. Development 136: 3699–3714, 2009. [DOI] [PubMed] [Google Scholar]
- 29.Myllarniemi M, Vuorinen K, Pulkkinen V, Kankaanranta H, Aine T, Salmenkivi K, Keski-Oja J, Koli K, Kinnula V. Gremlin localization and expression levels partially differentiate idiopathic interstitial pneumonia severity and subtype. J Pathol 214: 456–463, 2008. [DOI] [PubMed] [Google Scholar]
- 30.Nagaoka T, Morio Y, Casanova N, Bauer N, Gebb S, McMurtry I, Oka M. Rho/Rho kinase signaling mediates increased basal pulmonary vascular tone in chronically hypoxic rats. Am J Physiol Lung Cell Mol Physiol 287: L665–L672, 2004. [DOI] [PubMed] [Google Scholar]
- 31.Oka M, Morris KG, McMurtry IF. NIP-121 is more effective than nifedipine in acutely reversing chronic pulmonary hypertension. J Appl Physiol (1985) 75: 1075–1080, 1993. [DOI] [PubMed] [Google Scholar]
- 32.Otsuka F, Moore RK, Iemura S, Ueno N, Shimasaki S. Follistatin inhibits the function of the oocyte-derived factor BMP-15. Biochem Biophys Res Commun 289: 961–966, 2001. [DOI] [PubMed] [Google Scholar]
- 33.Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29: e45, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Shimoda LA, Sylvester JT, Sham JS. Mobilization of intracellular Ca2+ by endothelin-1 in rat intrapulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 278: L157–L164, 2000. [DOI] [PubMed] [Google Scholar]
- 35.Sidis Y, Mukherjee A, Keutmann H, Delbaere A, Sadatsuki M, Schneyer A. Biological activity of follistatin isoforms and follistatin-like-3 is dependent on differential cell surface binding and specificity for activin, myostatin, and bone morphogenetic proteins. Endocrinology 147: 3586–3597, 2006. [DOI] [PubMed] [Google Scholar]
- 36.Simeone-Penney MC, Severgnini M, Rozo L, Takahashi S, Cochran BH, Simon AR. PDGF-induced human airway smooth muscle cell proliferation requires STAT3 and the small GTPase Rac1. Am J Physiol Lung Cell Mol Physiol 294: L698–L704, 2008. [DOI] [PubMed] [Google Scholar]
- 37.Simon AR, Takahashi S, Severgnini M, Fanburg BL, Cochran BH. Role of the JAK-STAT pathway in PDGF-stimulated proliferation of human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 282: L1296–L1304, 2002. [DOI] [PubMed] [Google Scholar]
- 38.Song B, Jin H, Yu X, Zhang Z, Yu H, Ye J, Xu Y, Zhou T, Oudit GY, Ye JY, Chen C, Gao P, Zhu D, Penninger JM, Zhong JC. Angiotensin-converting enzyme 2 attenuates oxidative stress and VSMC proliferation via the JAK2/STAT3/SOCS3 and profilin-1/MAPK signaling pathways. Regul Pept 185: 44–51, 2013. [DOI] [PubMed] [Google Scholar]
- 39.Wang J, Shimoda LA, Sylvester JT. Capacitative calcium entry and TRPC channel proteins are expressed in rat distal pulmonary arterial smooth muscle. Am J Physiol Lung Cell Mol Physiol 286: L848–L858, 2004. [DOI] [PubMed] [Google Scholar]
- 40.Wang J, Weigand L, Lu W, Sylvester JT, Semenza GL, Shimoda LA. Hypoxia inducible factor 1 mediates hypoxia-induced TRPC expression and elevated intracellular Ca2+ in pulmonary arterial smooth muscle cells. Circ Res 98: 1528–1537, 2006. [DOI] [PubMed] [Google Scholar]
- 41.Wang J, Yang K, Xu L, Zhang Y, Lai N, Jiang H, Zhong N, Ran P, Lu W. Sildenafil inhibits hypoxia-induced transient receptor potential canonical protein expression in pulmonary arterial smooth muscle via cGMP-PKG-PPARgamma axis. Am J Respir Cell Mol Biol 49: 231–240, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wozney JM, Rosen V, Celeste AJ, Mitsock LM, Whitters MJ, Kriz RW, Hewick RM, Wang EA. Novel regulators of bone formation: molecular clones and activities. Science 242: 1528–1534, 1988. [DOI] [PubMed] [Google Scholar]
- 43.Yang J, Li X, Al-Lamki RS, Southwood M, Zhao J, Lever AM, Grimminger F, Schermuly RT, Morrell NW. Smad-dependent and smad-independent induction of id1 by prostacyclin analogs inhibits proliferation of pulmonary artery smooth muscle cells in vitro and in vivo. Circ Res 107: 252–262, 2010. [DOI] [PubMed] [Google Scholar]
- 44.Yang X, Long L, Southwood M, Rudarakanchana N, Upton PD, Jeffery TK, Atkinson C, Chen H, Trembath RC, Morrell NW. Dysfunctional Smad signaling contributes to abnormal smooth muscle cell proliferation in familial pulmonary arterial hypertension. Circ Res 96: 1053–1063, 2005. [DOI] [PubMed] [Google Scholar]
- 45.Yao Y, Jumabay M, Wang A, Bostrom KI. Matrix Gla protein deficiency causes arteriovenous malformations in mice. J Clin Invest 121: 2993–3004, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yao Y, Zebboudj AF, Shao E, Perez M, Bostrom K. Regulation of bone morphogenetic protein-4 by matrix GLA protein in vascular endothelial cells involves activin-like kinase receptor 1. J Biol Chem 281: 33921–33930, 2006. [DOI] [PubMed] [Google Scholar]
- 47.Yndestad A, Larsen KO, Oie E, Ueland T, Smith C, Halvorsen B, Sjaastad I, Skjonsberg OH, Pedersen TM, Anfinsen OG, Damas JK, Christensen G, Aukrust P, Andreassen AK. Elevated levels of activin A in clinical and experimental pulmonary hypertension. J Appl Physiol (1985) 106: 1356–1364, 2009. [DOI] [PubMed] [Google Scholar]
- 48.Zebboudj AF, Imura M, Bostrom K. Matrix GLA protein, a regulatory protein for bone morphogenetic protein-2. J Biol Chem 277: 4388–4394, 2002. [DOI] [PubMed] [Google Scholar]
- 49.Zimmerman LB, De Jesus-Escobar JM, Harland RM. The Spemann organizer signal noggin binds and inactivates bone morphogenetic protein 4. Cell 86: 599–606, 1996. [DOI] [PubMed] [Google Scholar]






