Propranolol represses infantile hemangioma cell growth through the β2-adrenergic receptor in a HIF-1α-dependent manner

  • Authors:
    • Peng Li
    • Zhengtuan Guo
    • Ya Gao
    • Weikang Pan
  • View Affiliations

  • Published online on: April 14, 2015     https://doi.org/10.3892/or.2015.3911
  • Pages: 3099-3107
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Propranolol, as a non-selective blocker of the β-adrenergic receptor (AR), is utilised as the first-line treatment for infantile hemangiomas. However, the underlying mechanism remains poorly understood. The present study was designed to investigate the molecular basis of propranolol on the regression of infantile hemangiomas using a proliferating infantile hemangioma-derived endothelial cell line. In infantile hemangioma patients, we found that propranolol significantly decreased the expression levels of the hypoxia inducible factor (HIF)-1α in serum and urine, as well as in hemangioma tissues. In vitro analysis revealed that propranolol reduces the expression of HIF-1α in hemangioma cells in a dose- and time-dependent manner, mainly by acting on β2-AR. Interestingly, it was observed that overexpression of HIF-1α apparently abrogated the inhibitory effects of propranolol on vascular endothelial growth factor (VEGF) expression and cell growth. Our data further demonstrated that propranolol inhibited the signal transducer and activator of transcription 3 (STAT3), a critical oncogenic signaling molecule, and the anti-apoptotic protein Bcl-2. Additionally, overexpression of HIF-1α significantly reversed the inhibitory effects of propranolol on STAT3 signaling. In a mouse xenograft hemangioma model, overexpression of HIF-1α significantly attenuated the therapeutic effects of propranolol and inhibited propranolol-induced hemangioma cell apoptosis. Moreover, the protein levels of VEGF, phosphorylated STAT3, total STAT3 and Bcl-2 were significantly upregulated by HIF-1α overexpression in propranolol-treated nude mice bearing hemangiomas. Collectively, our data provide evidence that propranolol may regress infantile hemangiomas by suppressing VEGF and STAT3 signaling pathways in an HIF-1α-dependent manner.

Introduction

Infantile hemangiomas are a type of benign tumors with high incidence in infancy (1). Typically, the lesions undergo a rapid proliferative phase during early infancy and gradually involute over the first few years (2). Although infantile hemangiomas are benign tumors and usually harmless, they can cause destruction and deformation of facial features, obstruction of breathing and vision, and even more life-threatening complications (35). However, the molecular pathogenesis of infantile hemangiomas remains largely unknown and effective treatment still needs to be developed.

Corticosteroids, as well as other drugs such as interferon α and vincristine, have been traditionally regarded as the first-line therapeutic approaches for the treatment of infantile hemangiomas (610). However, these drugs are accompanied by multiple and serious side-effects that are very painful for the infants (1113). Recently, a serendipitous discovery found that propranolol effectively regresses infantile hemangiomas when used to treat obstructive hypertrophic cardiomyopathy accompanied by an infantile hemangioma (14). Propranolol is widely used to treat infantile hemangiomas with satisfactory outcomes and no obviously serious side-effects (1519). Furthermore, infantile hemangiomas resistant to corticosteroids and interferon or severe infantile hemangiomas may be effectively treated by propranolol (20,21). It has been suggested that propranolol exerts its effects as a non-selective β-adrenergic receptor (AR) blocker that inhibits cell growth and induces cell apoptosis of the endothelial cells (21,22). However, the precise molecular mechanism of its action remains poorly understood.

In recent years, the role of β-AR in tumorigenesis has attracted increasing attention, which is associated with cell proliferation, apoptosis, invasion and metastasis of tumor cells (23). β-AR, including β1-AR and β2-AR, are G-protein-coupled receptors on endothelial cells that cause vasodilation of vessels upon activation (24). Moreover, activation of β-AR results in overexpression of the proangiogenic factors, including basic fibroblast growth factor and endothelial growth factor (VEGF) and inhibits cell apoptosis (21,22,25). It has been reported that β-AR inhibitors inhibit cell growth and angiogenesis and enhance cell apoptosis of tumors, thereby representing novel antitumor drugs (23). Hypoxia-inducible factor (HIF)-1 consists of two subunits, HIF-1α and HIF-1β, and plays an important role in regulating tumor progression (26,27). Of these two subunits, it has been suggested that HIF-1α is the major regulator for angiogenic factors such as VEGF (27). However, whether propranolol exerts its effects by regulating HIF-1α-mediated signaling needs to be further investigated.

In the present study, we speculated that β-AR-mediated and HIF-1α signaling may be associated with the therapeutic effects of propranolol in the treatment of infantile hemangiomas. We found that the elevated HIF-1α expression levels in infantile hemangioma patients were downregulated by propranolol. Using the hemangioma-derived endothelial cell line, propranolol was found to reduce the expression of HIF-1α in a dose- and time-dependent manner. Our results further demonstrated that propranolol inhibited HIF-1α expression through its action on β2-AR. Moreover, we revealed that propranolol suppressed cell growth by inhibiting HIF-1α-VEGF signaling. Additionally, the signal transducer and activator of transcription 3 (STAT3) and Bcl-2, which are critical oncogenic signaling molecules, were found to be increased in infantile hemangiomas, whereas propranolol was found to inhibit STAT3 and Bcl-2 expressions in a HIF-1α-dependent manner. Overexpression of HIF-1α attenuated the therapeutic effects of propranolol on hemangiomas in a mouse model, which further confirmed that propranolol regressed infantile hemangiomas in a HIF-1α-dependent manner. Collectively, we represent a potential mechanism of propranolol in which propranolol represses infantile hemangioma cell growth by inhibiting VEGF, STAT3 and Bcl-2 expression in a HIF-1α-dependent manner, which leads to cell growth arrest and the induction of cell apoptosis.

Materials and methods

Sample collections

A total of 11 infantile hemangioma patients were recruited in the present study from June 2012 to May 2013 at the Second Affiliated Hospital of Xi’an Jiaotong University. The patients were orally treated with propranolol (Tianjin Lisheng Pharmaceutical Co., Ltd., Tianjing, China). The patients had received no surgery or drug treatments prior to the propranolol treatment. Before and after the propranolol treatment, venous blood, urine and tumor tissues (d=2 mm) were collected with the informed consent of the family members and the approval of the Ethics Committee of the Second Affiliated Hospital of Xi’an Jiaotong University.

Animals and cell culture

Six-week-old female BAlB/c nude mice (20–30 g) were provided by the Experimental Animal Centre of the College of Medicine of Xi’an Jiaotong University. The mice were housed in a standard pathogen-free room according to the standard feeding protocols and the animal experimental procedures were handled in accordance with the Institutional Animal Care and Use Committee of Xi’an Jiaotong University. The hemangioma-derived endothelial cell line from proliferating infantile hemangioma tissues was previously prepared and established in our laboratory (28). The cell line was cultured in a RPMI-1640 medium (Invitrogen, Carlsbad, CA, USA) supplemented with 10 ng/ml epidermal growth factor and 15% fetal bovine serum (FBS) plus 100 U/ml penicillin and 100 μg/ml streptomycin in a humidified atmosphere chamber containing 5% CO2 at 37°C.

Enzyme-linked immunosorbent assay (ELISA)

The collected blood and urine samples were centrifuged at 1,000 x g for 15 min. The supernatants were collected and the concentration of HIF-1α was measured using an ELISA reagent kit (R&D Systems, Minneapolis, MN, USA) as per the supplier’s instructions and analyzed by an ELISA reader (BioTek Instruments Inc., Winooski, VT, USA).

Cell treatments and transfections

For propranolol treatment, hemangioma endothelial cells were treated with various concentrations of propranolol (0, 10, 50 and 100 μM) and incubated for 24, 48 and 72 h. For gene overexpression or gene knockdown, recombinant lentiviral vectors (Shanghai GenePharma Co., ltd., Shanghai, China) or specific siRNA (Santa Cruz Biotechnology, Santa Cruz, CA, USA) were transfected with the cells and incubated for 48 h prior to being collected for analysis.

Western blot analysis

Total proteins were extracted from tumor tissues or cells and quantified using a BCA protein assay kit (Thermo Fisher Scientific, Rockford, Il, USA). Approximately 25 μg protein was run on a precast 12.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis followed by electro-blotting onto a nitrocellulose membrane (Amersham, Little Chalfont, UK). The membranes were blocked by blocking buffer (3% skimmed milk solution) at 37°C for 1 h. The membranes were then blotted with primary antibodies diluted in blocking buffer overnight at 4°C. After being washed three times with Tris-buffered saline (TBS) and Tween (TBST) (each for 5 min), the membranes were incubated with horseradish peroxidase conjugated secondary antibody (Wuhan Boster Biological Technology, Ltd., Wuhan, China) in the blocking buffer for 1 h. The membranes were then washed three times with TBST and once with TBS, and the blots were developed with an enhanced chemiluminescence (ECL) detection system (Amersham). The following primary antibodies were used: anti-HIF-1α, anti-VEGF, anti-STAT3, anti-p-STAT3, anti-β1-AR, anti-β2-AR, anti-Bcl-2 and anti-GAPDH (Santa Cruz Biotechnology).

MTT assay

Cell growth was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Briefly, the cells were grown in 96-well plates (1×104 cells/well) and cultured until they reached 80% confluence. Thereafter, the cells were treated with HIF-1α, siRNA or lV-HIF-1α with 50 μM propranolol and incubated for 48 h. MTT diluted in PBS (5 mg/ml) was added at 20 μl/well and continually incubated for 4 h. Dimethylsulfoxide (150 μl/well) was added to dissolve the formazan crystals. The optical density value was measured using an ELISA reader at 490 nm.

Mouse xenograft experiment

A xenograft of hemangioma cells in the mice was prepared as previously described (28). Briefly, the cells (4×1010) that were diluted in 200 μl PBS were injected subcutaneously into the right groin. Two days after the tumor cell implantation, recombinant lentivirus (5×1010 plaque-forming units) in 200 μl PBS was injected subcutaneously into the left groin and this was performed every two weeks. The nude mice were intragastrically administered with propranolol solution (0.25 mg/ml) at 0.2 ml/10 g body weight every two days. After 40 days, the mice were euthanized by subcutaneous injection with sodium pentobarbital (40 mg/kg) and tumor tissues were harvested for analysis.

Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay

Cell apoptosis was detected by a TUNEL staining assay using a one-step TUNEL apoptosis assay kit (Beyotime, Haimen, China). Briefly, tumor tissues were dissected, fixed with 4% paraformaldehyde for 24 h at 4°C, gradient-dehydrated, embedded in paraffin and then serially cut into 5 μm-thick sections. The sections were dewaxed, gradient-rehydrated and digested with 20 μM proteinase for 10 min. After being washed with PBS, the apoptotic cells were examined using a TUNEl apoptosis assay kit according to the supplier’s instructions. The sections were observed under a fluorescence microscope (Olympus, Tokyo, Japan) and the TUNEl staining cells were calculated in five random fields and averaged per field.

Statistical analysis

The data were expressed as mean ± standard deviation (SD). Statistical analyses were performed using one-way ANOVA followed by the Bonferroni post hoc test among multiple groups. A value of P<0.05 was regarded as statistically significant.

Results

HIF-1α is downregulated by propranolol treatment in infantile hemangioma patients

To explore the molecular basis of propranolol in the treatment of infantile hemangioma, we evaluated the effect of propranolol on the levels of HIF-1α in infantile hemangioma patients. By using ELISA detection methods, we found that the concentrations of HIF-1α in the serum and urine were significantly increased in infantile hemangioma patients compared with those from healthy subjects. Surprisingly, the serum and urine levels of HIF-1α were significantly decreased after treatment with propranolol (Fig. 1A and B). To further validate that propranolol regulated the expression of HIF-1α, we detected the expression of HIF-1α in the hemangioma tumor tissues by western blot analysis. Similar results were obtained showing that propranolol treatment significantly decreased the protein levels of HIF-1α, which had been upregulated in infantile hemangioma tissues without treatment (Fig. 1C and D). Collectively, the results suggested that propranolol inhibited HIF-1α in infantile hemangioma patients.

Propranolol reduces the expression of HIF-1α in infantile hemangioma cells in a dose- and time-dependent manner

To further verify the regulated effect of propranolol on HIF-1α expression, we used the hemangioma-derived endothelial cell line to investigate the effect of propranolol on HIF-1α expression in vitro. Hemangioma endothelial cells were treated with varying concentrations of propranolol (0, 10, 50, 100 and 200 μM) for 48 h and the HIF-1α protein expression was determined by western blot analysis. The results showed that propranolol administration decreased the HIF-1α expression in a dose-dependent manner (Fig. 2A and B). Next, we treated the cells with 100 μM propranolol and incubated them for 24, 48 and 72 h (Fig. 2C and D). The results showed that HIF-1α expression was increased at 24 h and continuously increased up to 72 h. In conclusion, these results further indicated that propranolol regulated the expression of HIF-1α in the infantile hemangioma.

Propranolol inhibits the expression of HIF-1α mainly through acting on the β2-adrenergic receptor (AR)

Propranolol is a non-selective antagonist for β1-AR and β2-AR (21). To determine which β-AR played an important role in mediating the inhibitory effect of propranolol on the expression of HIF-1α, we added the β1-AR agonist (dobutamine) or the β2-AR agonist (salbutamol) to propranolol-treated cells and examined their effects on the expression of HIF-1α. The results showed that the β1-AR agonist had no apparent effect on the propranolol-induced HIF-1α decrease, whereas the β2-AR agonist significantly reversed the inhibitory effects of propranolol on HIF-1α expression (Fig. 3A and B). Additionally, inhibition of β2-AR by specific β2-AR siRNA had the same effect as propranolol on HIF-1α. However, knockdown of β1-AR had no apparent effect on HIF-1α expression (Fig. 3C and D). Collectively, these results suggested that propranolol repressed the expression of HIF-1α mainly through acting on β2-AR, not β1-AR.

Overexpression of HIF-1α abrogates the inhibitory effects of propranolol on hemangioma cell growth

To gain insight into HIF-1α in propranolol-induced cell growth arrest, we examined the effect of HIF-1α overexpression on propranolol-treated cells. The results showed that propranolol treatment inhibited the protein expression of HIF-1α and VEGF, whereas overexpression of HIF-1α blocked the inhibitory effects of propranolol on VEGF expression (Fig. 4A and B). Furthermore, propranolol-induced cell growth arrest was also abrogated by HIF-1α overexpression (Fig. 4C). Similarly, knockdown of HIF-1α resulted in a decreased VEGF expression (Fig. 4D and E) and cell growth inhibition (Fig. 4F), which had the same effect as propranolol treatment. These results implied that propranolol suppressed hemangioma cell growth by regulating HIF-1α.

Propranolol inhibits the expression of STAT3 and Bcl-2 in hemangioma cells

STAT3, a critical molecule for regulating oncogenic signaling, has been previously reported to be overexpressed in proliferating infantile hemangiomas (29). To investigate whether propranolol had an inhibitory effect on the expression of STAT3, we detected the protein expression of STAT3 in propranolol-treated hemangioma cells by western blot analysis. The results showed that different concentrations of propranolol significantly inhibited the expression of total STAT3 and phosphorylated STAT3 (p-STAT3). Furthermore, the downstream gene Bcl-2, an anti-apoptotic gene, was also markedly decreased by propranolol (Fig. 5). These data indicated that propranolol was capable of inhibiting STAT3 signaling.

Overexpression of HIF-1α abrogates the inhibitory effects of propranolol on STAT3 signaling

To investigate whether HIF-1α was involved in the regulation of STAT3 signaling, we detected the effect of HIF-1α overexpression on STAT3 signaling activation in propranolol-treated cells by western blot analysis (Fig. 6A). The results showed that overexpression of HIF-1α significantly abrogated the inhibitory effects of propranolol on the protein expression of p-STAT3, STAT3 and Bcl-2 (Fig. 6B). Knockdown of HIF-1α by siRNA markedly decreased the protein expression of p-STAT3, STAT3 and Bcl-2, which mimics the effect of propranolol on STAT3 signaling. Collectively, these results suggested that HIF-1α played an important role in propranolol-mediated inhibition of STAT3 signaling in hemangioma cells.

Overexpression of HIF-1α reduces the therapeutic effects of propranolol on hemangiomas in a mouse model

To further validate the important role of HIF-1α involved in propranolol treatment for hemangiomas, we infected a mouse xenograft hemangioma model with lV-HIF-1α overexpressing HIF-1α. Using the TUNEL method, we found that propranolol-induced hemangioma apoptosis was apparently inhibited by HIF-1α overexpression (Fig. 7A). Furthermore, the protein expression of VEGF, p-STAT3, STAT3 and Bcl-2, downregulated by propranolol in tumor tissues, was significantly upregulated by HIF-1α overexpression (Fig. 7B and C). Collectively, these results further confirmed that propranolol repressed hemangiomas in a HIF-1α-dependent manner.

Discussion

Although the use of propranolol has been broadly applied in the clinical treatment of infantile hemangiomas, the underlying mechanism remains largely unknown. Therefore, it is of great importance to gain better insight into propranolol in the treatment of infantile hemangiomas to enable better patient care as well as to contribute to the development of novel therapies. In the present study, we sought to delineate the molecular mechanism of propranolol in the treatment of infantile hemangiomas. Furthermore, evidence was presented that propranolol represses infantile hemangioma cell growth through its action on β2-AR and inhibits VEGF, STAT3 and Bcl-2 expression in an HIF-1α-dependent manner, which leads to cell growth arrest and the induction of cell apoptosis using a hemangioma endothelial cell model.

Formerly, propranolol was generally used for the treatment of cardiovascular diseases including hypertension, supraventricular tachycardia, ischemic heart disease and arrhythmia, and has been proved to be safe and well tolerated (30). In 2008 Léauté-Labrèze et al (14) used propranolol to treat obstructive hypertrophic cardiomyopathy accompanied by infantile hemangiomas and unexpectedly found that propranolol effectively regresses infantile hemangiomas. Since then, an increasing number of studies have reported the use of propranolol in the treatment of infantile hemangiomas (3133). Currently, propranolol is regarded as the first-line drug for the treatment of infantile hemangiomas due to its capacity to provide instant gratification and be more effective, and its few side-effects, as well as its low cost (3436). However, the underlying mechanism is still elusive. In recent years, numerous studies have been devoted to investigating the molecular basis of propranolol in the treatment of infantile hemangiomas. Some possible mechanisms have been proposed, including vasoconstriction, decreased expression of VEGF and the triggering of apoptosis (22). Nonetheless, there is still a lack of precise understanding.

β-AR has been reported to be expressed in various tumor cells and the activation of β-AR results in an increase in the cyclic AMP (cAMP) that activates the cAMP-dependent protein kinases and the multiple signaling pathways (3739). Many studies have demonstrated that activation of β-AR exhibits the tumor-promoting function in various tumor cells (40,41). β-AR antagonists have been found to inhibit cell proliferation, invasion and migration of the tumor cells (4244). Propranolol has been reported as a non-selective β-AR blocker of β1-AR and β2-AR (21). In the present study, we found that propranolol inhibited cell growth of hemangioma endothelial cells mainly through its action on β2-AR, and knockdown of β2-AR had the same effect as propranolol on cell growth, while knockdown of β1-AR had no apparent effect on cell growth. Our results indicated that propranolol repressed infantile hemangiomas by inhibiting β2-AR-mediated signaling pathways. Consistently, Truong et al (45) reported that β2-AR expression and phosphorylation responded to propranolol treatment of infantile hemangiomas.

HIF-1α has been proposed as an important mediator in regulating tumor progression (26,27). In the present study, we found that HIF-1α was upregulated in the serum, urine and tumor tissues in infantile hemangioma patients, and treatment of propranolol markedly inhibited the expression levels of HIF-1α. The results also implied that HIF-1α was involved in regulating infantile hemangiomas. Our data further identified that propranolol suppressed HIF-1α by inhibiting β2-AR. The link between HIF-1α and β-AR has been reported in a variety of tumors, including breast, prostate and pancreatic cancer cells (46,47). β-AR agonists have been shown to increase HIF-1α expression, and β-AR agonist-induced HIF-1α and VEGF expression was abrogated by propranolol (47). More recently, propranolol was suggested to repress hemangioma cells by downregulating HIF-1α, VEGF and downstream signaling pathways (48). The present study presented evidence that propranolol blocks β2-AR leading to decreased HIF-1α and VEGF expression, which may explain the molecular basis of propranolol action. Furthermore, our data demonstrated that propranolol inhibited the expression and phosphorylation of STAT3 in infantile hemangioma cells. The overexpression of STAT3 has been reported in infantile hemangiomas (29) and angiosarcomas (49).

In summary, our results suggested that propranolol repressed infantile hemangioma cell growth through its action on β2-AR and inhibits VEGF, STAT3 and Bcl-2 expression leading to cell growth arrest and the induction of cell apoptosis using a hemangioma endothelial cell model. Propranolol treatment also decreased the expression of HIF-1α, which is a critical regulator of tumor progression. Overexpression of HIF-1α significantly abrogated the effects of propranolol, implying that propranolol regressed infantile hemangiomas in a HIF-1α-dependent manner. These findings suggested a plausible mechanism of propranolol in the regression of infantile hemangiomas and provided novel insights for the development of novel therapeutic methods for infantile hemangiomas.

Acknowledgments

The present study was supported by grants from the National Natural Science Foundation of China (no. 81172589).

Abbreviations:

AR

β-adrenergic receptor

HIF

hypoxia inducible factor

VEGF

vascular endothelial growth factor

STAT3

signal transducer and activator of transcription 3

TUNEL

terminal deoxynucleotidyl transferase dUTP nick-end labeling

Referencs

1 

Kilcline C and Frieden IJ: Infantile hemangiomas: How common are they? (A systematic review of the medical literature). Pediatr Dermatol. 25:168–173. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Chang LC, Haggstrom AN, Drolet BA, Baselga E, Chamlin SL, Garzon MC, Horii KA, Lucky AW, Mancini AJ, Metry DW, et al: Growth characteristics of infantile hemangiomas: Implications for management. Pediatrics. 122:360–367. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Leaute-Labreze C, Prey S and Ezzedine K: Infantile haemangioma: Part I. Pathophysiology, epidemiology, clinical features, life cycle and associated structural abnormalities. J Eur Acad Dermatol Venereol. 25:1245–1253. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Metry DW, Haggstrom AN, Drolet BA, Baselga E, Chamlin S, Garzon M, Horii K, Lucky A, Mancini AJ, Newell B, et al: A prospective study of PHACE syndrome in infantile hemangiomas: Demographic features, clinical findings, and complications. Am J Med Genet A. 140:975–986. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Pandey A, Gangopadhyay AN and Upadhyay VD: Evaluation and management of infantile hemangioma: An overview. Ostomy Wound Manage. 54:16–18. 2008.PubMed/NCBI

6 

Bennett ML, Fleischer AB Jr, Chamlin SL and Frieden IJ: Oral corticosteroid use is effective for cutaneous hemangiomas: An evidence-based evaluation. Arch Dermatol. 137:1208–1213. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Ezekowitz RA, Mulliken JB and Folkman J: Interferon alfa-2a therapy for life-threatening hemangiomas of infancy. N Engl J Med. 326:1456–1463. 1992. View Article : Google Scholar : PubMed/NCBI

8 

Perez J, Pardo J and Gomez C: Vincristine - an effective treatment of corticoid-resistant life-threatening infantile hemangiomas. Acta Oncol. 41:197–199. 2002. View Article : Google Scholar

9 

Perez Payarols J, Pardo Masferrer J and Gomez Bellvert C: Treatment of life-threatening infantile hemangiomas with vincristine. N Engl J Med. 333:691995. View Article : Google Scholar : PubMed/NCBI

10 

Moore J, Lee M, Garzon M, Soffer S, Kim E, Saouaf R, del Toro G, Yamashiro D and Kandel J: Effective therapy of a vascular tumor of infancy with vincristine. J Pediatr Surg. 36:1273–1276. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Boon LM, MacDonald DM and Mulliken JB: Complications of systemic corticosteroid therapy for problematic hemangioma. Plast Reconstr Surg. 104:1616–1623. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Dubois J, Hershon L, Carmant L, Belanger S, Leclerc JM and David M: Toxicity profile of interferon alfa-2b in children: A prospective evaluation. J Pediatr. 135:782–785. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Barlow CF, Priebe CJ, Mulliken JB, Barnes PD, MacDonald D, Folkman J and Ezekowitz RA: Spastic diplegia as a complication of interferon Alfa-2a treatment of hemangiomas of infancy. J Pediatr. 132:527–530. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Léauté-Labrèze C, Dumas de la Roque E, Hubiche T, Boralevi F, Thambo JB and Taieb A: Propranolol for severe hemangiomas of infancy. N Engl J Med. 358:2649–2651. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Bigorre M, Van Kien AK and Valette H: Beta-blocking agent for treatment of infantile hemangioma. Plast Reconstr Surg. 123:195e–196e. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Holmes WJ, Mishra A, Gorst C and Liew SH: Propranolol as first-line treatment for rapidly proliferating infantile haemangiomas. J Plast Reconstr Aesthet Surg. 64:445–451. 2011. View Article : Google Scholar

17 

Buckmiller LM, Munson PD, Dyamenahalli U, Dai Y and Richter GT: Propranolol for infantile hemangiomas: Early experience at a tertiary vascular anomalies center. Laryngoscope. 120:676–681. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Solman L, Murabit A, Gnarra M, Harper JI, Syed SB and Glover M: Propranolol for infantile haemangiomas: Single centre experience of 250 cases and proposed therapeutic protocol. Arch Dis Child. 99:1132–1136. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Baetz J, Eigelshoven S, Marquard J, Bruch-Gerharz D, Homey B and Meissner T: Infantile hemangioma. Successful treatment with propranolol. Hautarzt. 61:290–292. 2010.In German. View Article : Google Scholar : PubMed/NCBI

20 

Sarialioglu F, Erbay A and Demir S: Response of infantile hepatic hemangioma to propranolol resistant to high-dose methylprednisolone and interferon-α therapy. Pediatr Blood Cancer. 55:1433–1434. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Storch CH and Hoeger PH: Propranolol for infantile haemangiomas: Insights into the molecular mechanisms of action. Br J Dermatol. 163:269–274. 2010. View Article : Google Scholar : PubMed/NCBI

22 

D’Angelo G, Lee H and Weiner RI: cAMP-dependent protein kinase inhibits the mitogenic action of vascular endothelial growth factor and fibroblast growth factor in capillary endothelial cells by blocking Raf activation. J Cell Biochem. 67:353–366. 1997. View Article : Google Scholar

23 

Perez-Sayans M, Somoza-Martin JM, Barros-Angueira F, Diz PG, Gandara Rey JM and Garcia-Garcia A: Beta-adrenergic receptors in cancer: therapeutic implications. Oncol Res. 19:45–54. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Guimaraes S and Moura D: Vascular adrenoceptors: An update. Pharmacol Rev. 53:319–356. 2001.PubMed/NCBI

25 

Iaccarino G, Ciccarelli M, Sorriento D, Galasso G, Campanile A, Santulli G, Cipolletta E, Cerullo V, Cimini V, Altobelli GG, et al: Ischemic neoangiogenesis enhanced by beta2-adrenergic receptor overexpression: A novel role for the endothelial adrenergic system. Circ Res. 97:1182–1189. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Semenza GL: Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu Rev Cell Dev Biol. 15:551–578. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Li P, Xiao X, Xu Q and Guo Z: Establishment of human infancy hemangioma-derived endothelial cell line XPTS-1 and animal model of human infancy hemangioma. Zhonghua Kou Qiang Yi Xue Za Zhi. 46:129–133. 2011.In Chinese. PubMed/NCBI

29 

Itinteang T, Tan ST, Brasch HD, Steel R, Best HA, Vishvanath A, Jia J and Day DJ: Infantile haemangioma expresses embryonic stem cell markers. J Clin Pathol. 65:394–398. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Loaldi A, Polese A, Montorsi P, De Cesare N, Fabbiocchi F, Ravagnani P and Guazzi MD: Comparison of nifedipine, propranolol and isosorbide dinitrate on angiographic progression and regression of coronary arterial narrowings in angina pectoris. Am J Cardiol. 64:433–439. 1989. View Article : Google Scholar : PubMed/NCBI

31 

Harikrishna B, Ganesh A, Al-Zuahibi S, Al-Jabri S, Al-Waily A, Al-Riyami A, Al-Azri F, Masoud F and Al-Mujaini A: Oral propranolol for the treatment of periorbital infantile hemangioma: A preliminary report from Oman. Middle East Afr J Ophthalmol. 18:298–303. 2011. View Article : Google Scholar

32 

Manunza F, Syed S, Laguda B, Linward J, Kennedy H, Gholam K, Glover M, Giardini A and Harper JI: Propranolol for complicated infantile haemangiomas: A case series of 30 infants. Br J Dermatol. 162:466–468. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Solomon T, Ninnis J, Deming D, Merritt TA and Hopper A: Use of propranolol for treatment of hemangiomas in PHACE syndrome. J Perinatol. 31:739–741. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Price CJ, Lattouf C, Baum B, McLeod M, Schachner LA, Duarte AM and Connelly EA: Propranolol vs corticosteroids for infantile hemangiomas: A multicenter retrospective analysis. Arch Dermatol. 147:1371–1376. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Bertrand J, McCuaig C, Dubois J, Hatami A, Ondrejchak S and Powell J: Propranolol versus prednisone in the treatment of infantile hemangiomas: A retrospective comparative study. Pediatr Dermatol. 28:649–654. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Lou Y, Peng WJ, Cao Y, Cao DS, Xie J and Li HH: The effectiveness of propranolol in treating infantile haemangiomas: a meta-analysis including 35 studies. Br J Clin Pharmacol. 78:44–57. 2014. View Article : Google Scholar

37 

Montminy M: Transcriptional regulation by cyclic AMP. Annu Rev Biochem. 66:807–822. 1997. View Article : Google Scholar : PubMed/NCBI

38 

Zhang X, Odom DT, Koo SH, Conkright MD, Canettieri G, Best J, Chen H, Jenner R, Herbolsheimer E, Jacobsen E, et al: Genome-wide analysis of cAMP-response element binding protein occupancy, phosphorylation, and target gene activation in human tissues. Proc Natl Acad Sci USA. 102:4459–4464. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Cole SW and Sood AK: Molecular pathways: beta-adrenergic signaling in cancer. Clin Cancer Res. 18:1201–1206. 2012. View Article : Google Scholar :

40 

Powe DG and Entschladen F: Targeted therapies: Using beta-blockers to inhibit breast cancer progression. Nat Rev Clin Oncol. 8:511–512. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Tang J, Li Z, Lu L and Cho CH: beta-Adrenergic system, a backstage manipulator regulating tumour progression and drug target in cancer therapy. Semin Cancer Biol. 23:533–542. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Shi M, Yang Z, Hu M, Liu D, Hu Y, Qian L, Zhang W, Chen H, Guo L, Yu M, et al: Catecholamine-induced β2-adrenergic receptor activation mediates desensitization of gastric cancer cells to trastuzumab by upregulating MUC4 expression. J Immunol. 190:5600–5608. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Entschladen F, Drell Tl IV, Lang K, Joseph J and Zaenker KS: Tumour-cell migration, invasion, and metastasis: navigation by neurotransmitters. Lancet Oncol. 5:254–258. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Sood AK, Bhatty R, Kamat AA, Landen CN, Han L, Thaker PH, Li Y, Gershenson DM, Lutgendorf S and Cole SW: Stress hormone-mediated invasion of ovarian cancer cells. Clin Cancer Res. 12:369–375. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Truong MT, Chang KW, Berk DR, Heerema-McKenney A and Bruckner AL: Propranolol for the treatment of a life-threatening subglottic and mediastinal infantile hemangioma. J Pediatr. 156:335–338. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Hu HT, Ma QY, Zhang D, Shen SG, Han L, Ma YD, Li RF and Xie KP: HIF-1alpha links beta-adrenoceptor agonists and pancreatic cancer cells under normoxic condition. Acta Pharmacol Sin. 31:102–110. 2010. View Article : Google Scholar

47 

Park SY, Kang JH, Jeong KJ, Lee J, Han JW, Choi WS, Kim YK, Kang J, Park CG and Lee HY: Norepinephrine induces VEGF expression and angiogenesis by a hypoxia-inducible factor-1alpha protein-dependent mechanism. Int J Cancer. 128:2306–2316. 2011. View Article : Google Scholar

48 

Chim H, Armijo BS, Miller E, Gliniak C, Serret MA and Gosain AK: Propranolol induces regression of hemangioma cells through HIF-1alpha-mediated inhibition of VEGF-A. Ann Surg. 256:146–156. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Lin N, Uchi H, Moroi Y, Fukiwake N, Dainichi T, Takeuchi S, Takahara M, Tu Y, Furue M and Urabe K: Significance of the expression of phosphorylated signal transducer and activator of transcription-3, -Akt, and -cyclin D1 in angiosarcoma. J Dermatol Sci. 48:64–66. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 33 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li P, Guo Z, Gao Y and Pan W: Propranolol represses infantile hemangioma cell growth through the β2-adrenergic receptor in a HIF-1α-dependent manner. Oncol Rep 33: 3099-3107, 2015
APA
Li, P., Guo, Z., Gao, Y., & Pan, W. (2015). Propranolol represses infantile hemangioma cell growth through the β2-adrenergic receptor in a HIF-1α-dependent manner. Oncology Reports, 33, 3099-3107. https://doi.org/10.3892/or.2015.3911
MLA
Li, P., Guo, Z., Gao, Y., Pan, W."Propranolol represses infantile hemangioma cell growth through the β2-adrenergic receptor in a HIF-1α-dependent manner". Oncology Reports 33.6 (2015): 3099-3107.
Chicago
Li, P., Guo, Z., Gao, Y., Pan, W."Propranolol represses infantile hemangioma cell growth through the β2-adrenergic receptor in a HIF-1α-dependent manner". Oncology Reports 33, no. 6 (2015): 3099-3107. https://doi.org/10.3892/or.2015.3911