Abstract
Infantile hemangioma (IH), a benign vascular tumor, is the most common tumor of infancy, with an incidence of 5%–10% at the end of the first year. The tumor displays a distinctive life cycle consisting of a proliferating phase, occurring in the first months of life, followed by an involuting phase. Thus, IH represents a unique model of postnatal vasculogenesis, angiogenesis, and vessel regression. Traditionally, corticosteroids were the drug of choice when treatment of IH was indicated. In recent years, beta-blockers, most specifically propranolol, have serendipitously been shown to be an effective pharmacological treatment. This article will focus on the mechanism of action of these two drugs, the old and the new treatments, in slowing the growth and accelerating involution of IH.
Propranolol has rapidly emerged as a new first-line treatment for endangering infantile hemangioma. Identifying its mechanism of action may aid in developing target-specific antiangiogenic cancer therapies.
Infantile hemangioma (IH), a benign vascular tumor, is the most common tumor of infancy, with an incidence of 5%–10% at the end of the first year. There is increased risk in premature neonates under the weight of 1500 g (Amir et al. 1986), in females and in Caucasians (Haggstrom et al. 2007). The tumor displays a distinctive life cycle that can be separated, both clinically and histologically, into three phases (Enjolras and Mulliken 1993; Frieden et al. 2005). The proliferating phase starts within a few weeks from birth and ends within the first year of life, with the most growth occurring during the first 4–6 months of life (Fig. 1) (Chang et al. 2008). In the proliferating phase the tumor is composed of densely packed cells that express endothelial and pericytic markers, with features of immature vessels such as plump endothelium and high nuclei/cytoplasmic ratio. Multipotent progenitor-like cells reside outside of the vessels (Khan et al. 2008; Boscolo and Bischoff 2009). The involuting phase typically begins at 1 yr of age. Clinically, the infantile hemangioma shrinks centrifugally from the center of the lesion, changes color to less red, and softens. The vascular channels appear more mature, with flattened endothelial cells, an organized perivascular layer, and basement membrane. There is a prominent apoptosis, some occurring in endothelial cells (Iwata et al. 1996; Razon et al. 1998; Dosanjh et al. 2000), and an increase in the number of mast cells. Finally, at the involuted phase, tumor growth has stopped and the tumor has regressed. The regression is either complete or, more often, leaves scar tissue, telangiectasia, or redundant or anetodermic skin (Jackson 1998). When viewed in histological sections, fat, fibroblasts, and connective tissue replace the vascular tissue at this stage, with few large feeding and draining vessels evident.
IH is a benign tumor, and in most patients no specific treatment is required. However, in 10% of cases IH is problematic or even endangering to the child, owing to its location or owing to excessive growth. In these circumstances that carry the risk of irreversible disfigurement, airway obstruction, or decreased vision, treatment is indicated. Corticosteroids have been the first-line treatment for IH for more than 40 years (Zarem and Edgerton 1967; Cohen and Wang 1972). In recent years, beta-blockers, most specifically propranolol, have serendipitously been shown to be an effective pharmacological treatment of proliferating IH. Other, less common medications are interferon-2α and vincristine (Maguiness and Frieden 2010). This article will focus on the mechanism of action of corticosteroids and propranolol, the old and the new treatments, in slowing the growth and accelerating involution of IH. For each drug, a general mechanism of action will be reviewed, then its specific mechanism against IH, and finally, broader mechanisms in antiangiogenic therapy.
CORTICOSTEROIDS
General Mechanism of Action
Glucocorticoids, natural or synthetic, belong to a class of steroid hormones that bind to the glucocorticoid receptor (GR). Unbound GR resides within the cytoplasm in an inactive state, as an oligomeric complex with regulatory proteins such as the HSP90 (heat shock protein-90 KD) (Dalman et al. 1989), HSP70 (heat shock protein-70 KD), and the p59 immunophilin. On diffusion of the glucocorticoid ligand into the cytoplasm and its binding to high-affinity GR, the receptor is activated, and a nuclear localization signal is unmasked (Pelaia et al. 2003). The GR then translocates to the nucleus where it binds to a glucocorticoid response element (GRE) within the controlling region for glucocorticoid responsive target genes (Adcock et al. 2006). Once bound to DNA, GR can elicit either transactivation or transrepression of gene expression. Transactivation occurs through the recruitment of complexes containing basal transcription factors, coactivators, chromatin modifiers, and RNA polymerase II, which together induce histone modifications and chromatin remodeling that lead to increased production of messenger RNAs (mRNAs) (Adcock et al. 2006). As for transrepression, GR can bind to a GRE that overlaps the DNA-binding site for a transcription factor or the start site of transcription, thus preventing gene expression. Importantly for IH, GR can repress AP-1/NF-kB-mediated gene expression. Furthermore, it can interact with corepressors such as NCoR (nuclear receptor corepressor receptor) or dephosphorylate RNA polymerase II (Schaaf and Cidlowski 2002; Schoneveld et al. 2004). Finally, GR can induce rapid nongenomic effects through a membrane-associated receptor (Norman et al. 2004) or by modulating the activity of kinases such as ERK, Akt, or phosphatidylinositol 3-kinase and protein kinase Akt.
Mechanism of Action in Infantile IH
When studying the effect of a drug on a tumoral process, one has to consider the activity on the different cellular compartments as well as on the inter-relationships between the cells. Thus, a brief description of the major cell types in IH is provided. Endothelial cells constitute about 20% of the cell population within the tumor (Yu et al. 2004). These hemangioma-derived endothelial cells have been shown to be clonal, have immature morphology, and display increased growth and migratory properties compared to normal dermal endothelial cells (Dosanjh et al. 2000; Boye et al. 2001; Yu et al. 2001; Dadras et al. 2004). Pericytes, or mural cells, are arranged around the endothelial cell layer in a proliferating IH tissue section (Li et al. 2003; Boscolo and Bischoff 2009). Several cell types have been identified in the tumor’s interstitium. Among them are mast cells, which increase in number during involution (Qu et al. 1995; Tan et al. 2004), and myeloid cells (CD45+/CD14+), which are present during the proliferative phase. Hemangioma progenitor cells or hemangioma-derived stem cells (HemSCs) have been identified and isolated by us based on the expression of the human stem/progenitor cell marker CD133 (Shmelkov et al. 2005). These cells have self-renewal capacity and display multipotential activity, being able to differentiate in vivo into endothelial cells and adipocytes (Khan et al. 2008). When injected subcutaneously into nude mice, HemSC form human blood vessels that express GLUT-1, a specific IH marker. Hence—the HemSC are vasculogenic—able to form blood vessels de novo. Furthermore, 2 mo after injection the vessels are replaced by fat tissue, indicating the HemSCs can recapitulate key features of the life cycle of IH.
Evidence from In Vitro and In Vivo Studies of IH
Blocking of the Vasculogenic Potential of HemSC
To study the effect of corticosteroids on vasculogenic processes that occur when HemSC are implanted in vivo, we used our animal model. HemSC were injected subcutaneously into nude mice, and dexamethasone (a synthetic glucocorticoid) was injected intraperitoneally thereafter for 7 d. Dexamethasone inhibited the formation of blood vessels in a dose-dependent manner (Greenberger et al. 2010b). Furthermore, by pretreating the HemSC and endothelial cells with dexamethasone, each separately in vitro and before implantation, we were able to show that HemSCs, and not the endothelial cells, are the target of corticosteroids. This indicated that steroids might work by inhibiting the vasculogenesis that occurs in IH.
Down-Regulation of Vascular Endothelial Growth Factor A
Vascular endothelial growth factor A (VEGF-A), a master regulator of angiogenesis and vasculogenesis, has been shown by us and by others to be present at higher levels in IH tumors in the proliferating phase compared to the involuting phase (Takahashi et al. 1994; Chang et al. 1999; Kleinman et al. 2007; Greenberger et al. 2010a,b). It has also been shown that corticosteroids suppress the expression of VEGF-A in a variety of cell types (Bandi and Kompella 2001; Kompella et al. 2003; Kim et al. 2008). We found that the corticosteroids dexamethasone, prednisone, prednisolone, methylprednisolone, and hydrocortisone were each able to dramatically down-regulate VEGF-A secretion by HemSC. The suppression was reversed by RU-486, a synthetic steroid hormone antagonist, indicating a GR-dependent mechanism (Greenberger et al. 2010b). These findings are consistent with reports that show decreased levels of VEGF-A in the serum of IH patients following systemic steroid therapy (Zhang et al. 2005). Silencing the expression of VEGF-A in HemSC by short hairpin RNA (shRNA) was sufficient to block blood vessel formation in vivo (Greenberger et al. 2010b). The effect of the steroids was not mediated by the induction of apoptosis as dexamethasone did not lead to reduced viability of HemSC or endothelial cells, even at concentrations 1000 times higher than those that led to VEGF suppression.
Modification of Proangiogenic Profile
The direct target genes of corticosteroids in vivo are still largely unknown and might be tissue or even cell specific (Phuc Le et al. 2005; van Batenburg et al. 2010). Examining HemSC and endothelial cells in vitro revealed that several corticosteroid-modified genes with relevance for angiogenesis, in addition to VEGF-A, were expressed (Greenberger et al. 2010b). Monocyte chemotactic protein-1 (MCP-1), which has been reported to be overexpressed in proliferating versus involuting infantile hemangioma (Isik et al. 1996), was suppressed by dexamethasone. Interleukin-6, which was detected in in vitro hemangioma models (Hasan et al. 2003), was also suppressed, as were matrix metalloproteinase-1 (MMP-1) and urokinase-type plasminogen activator receptor (uPAR). These target genes, MCP-1, uPAR, and IL-6, were found to be differentially expressed in proliferating versus involuting IH tissue by quantitative real-time polymerase chain reaction (PCR) measurements (Greenberger et al. 2010b). In sum, corticosteroids seem to affect key molecules in the angiogenesis process when analyzed in HemSC and hemangioma-derived endothelial cells cultured in vitro.
Suppression of Nuclear Factor κ-Light-Chain-Enhancer of Activated B Cells Activity
Corticosteroids are known to be regulators of nuclear factor κ-light-chain enhancer of activated B cells (NF-κB) activity, through several distinct mechanisms (Tao et al. 2001), for example, (1) physical interaction between NF-κB subunits and GR (Caldenhoven et al. 1995), (2) increased transcription of the NF-κB inhibitor IκBα (Auphan et al. 1995), and (3) competition for interactions with coactivators (Kamei et al. 1996; Lee et al. 1998). In HemSC, corticosteroids suppress NF-κB activity. This suppression in turn leads to down-regulation of VEGF-A, as well as other proangiogenic cytokines MCP-1, uPAR, MMP-1, and IL-6 (Greenberger et al. 2010a). As VEGF-A is critical for hemangiogenesis, this finding suggests that the effect of corticosteroids is to interfere with NF-κB regulation. In addition, suppression of NF-κB may also have direct antiendothelial effects, as NF-κB has been shown to contribute to embryonic carcinoma (EC) survival in vitro, and NF-κB blockade in vivo sensitizes ECs to stress-induced apoptosis (Kisseleva et al. 2006).
Antagonizing Estrogen’s Effects
Corticosteroids have been shown to antagonize estrogen responses (Bever et al. 1956; Rhen et al. 2003), including the prosurvival effects of estrogen (Zhou et al. 1989) and its stimulatory effects on insulin growth factor-1 (IGF-1) (Sahlin 1995). This inhibition might occur through corticosteroid induction of the expression and activity of estrogen sulfotransferase, an enzyme that deactivates estrogens (Gong et al. 2008). In infants with proliferating IH, serum levels of estrogen (estradiol 17β) have been shown to be increased compared to normal infants. (Sasaki et al. 1984; Liu et al. 1999). Furthermore, estrogen receptor (ER) expression and binding activity were reported to be higher in patients with proliferating IH (Sasaki et al. 1984; Kleinman et al. 2007) and to decrease following corticosteroid treatment (Sasaki et al. 1984). As estrogen has been shown to be essential for neoangiogenesis (Das et al. 2009) and vasculogenesis (Masuda et al. 2007) in the reproductive system, it might then be the case that corticosteroids exert an additional effect on IH—that of antagonizing the activity of estrogen.
PROPRANOLOL
Propranolol as a pharmacological treatment of IH was first reported in 2008, in two children who showed rapid regression of disease when treated with propranolol for cardiopulmonary indications (Leaute-Labreze et al. 2008). Since then, propranolol treatment for IH has become popular worldwide and positive results with its use are reported by many groups (Cheng et al. 2010; Mazereeuw-Hautier et al. 2010; Rosbe et al. 2010). Propranolol is a noncardioselective β-adrenergic receptor blocker used traditionally for other indications such as hypertension, angina pectoris, myocardial infarction, migraines, anxiety disorders, and tremor. The mechanism of action of propranolol on IH remains elusive. To date there are no studies on the effects of propranolol using IH animal models or cultured IH-derived cells. Therefore, we will summarize here the general mechanism of action of propranolol and suggest possible mechanisms based on data derived from studies performed using angiogenesis assays and endothelial cells.
General Mechanism of Action
When catecholamines, adrenaline and noradrenaline, interact with the β-adrenergic receptors β1 or β2, an agonist-promoted binding of the receptor to the heterotrimeric guanosine triphosphate-binding protein Gs occurs, causing dissociation of Gα-GTP and Gβγ subunits. This in turn leads to activation of adenylate cyclase and production of cyclic adenosine monophosphate (cAMP). Downstream effectors of cAMP include cAMP-dependent protein kinase (PKA) as well as cAMP-gated ion channels (Benovic 2002). Propranolol is an orthosteric antagonist of both β1- and β2-adrenergic receptors. In addition, it has been shown to function as central serotonin 5-HT receptor antagonist, inhibitor of noradrenaline reuptake and indirect agonist of α-adrenergic receptors (Young and Glennon 2009). The drug exists as a pair of optical isomers: S(−)propranolol and R(+)propranolol. The enantiomers bind with relatively large differences in affinity to the β-adrenoceptors (Young and Glennon 2009). Most commercial preparations contain a mix of the two isomers.
Hemodynamic Effects
Following the administration of propranolol, a rapid change in the tumor is typically noticed, including decreased redness and softening (Sans et al. 2009; Rosbe et al. 2010). This raises the question of whether propranolol exerts its effect via vasoconstriction of the high-flow blood vessels feeding the IH tumor. Propranolol has been shown to decrease blood flow to many tissues following single administration (Nies et al. 1973; McSorley and Warren 1978; Vandenburg et al. 1981). Particularly in the skin, adrenaline-induced vasoconstriction has been shown to be increased by oral propranolol (Doshi et al. 1984). However, as prolonged treatment probably does not affect vascular resistance in subcutaneous tissues (Jensen et al. 1983), it is reasonable to assume that there are additional mechanisms that account for the prolonged effects of propranolol on the course of IH.
VEGF-A
VEGF-A is a critical factor for the growth of IH. Thus, suppression of VEGF-A by propranolol is a plausible mechanism for its activity. Noradrenaline has been shown to enhance VEGF-A production by several cell types, both normal and cancerous (Fredriksson et al. 2000; Lutgendorf et al. 2003; Park et al. 2011). Furthermore, in an ovarian carcinoma animal model, catecholamines up-regulated tumoral VEGF and increased angiogenesis (Thaker et al. 2006). These effects of the catecholamines are mediated by the β1 and β2 adrenergic receptors and are blocked by propranolol (Fredriksson et al. 2000; Park et al. 2011). In addition to VEGF, noradrenaline up-regulates HIF-1α protein (Park et al. 2011). The signal transduction pathway leading to HIF-1α and VEGF-A up-regulation is mediated through cAMP and PKA (Fredriksson 2000). In some cell lines, Src tyrosine kinase, a downstream effector of PKA, is involved, however, not through activation of the mitogen-activated protein (MAP) kinases Erk1/2 (Fredriksson et al. 2000). Importantly, propranolol does not affect baseline level of VEGF-A production by the cells, but rather opposes catecholamine stimulation (Fredriksson et al. 2000; Thaker et al. 2006). Thus, if VEGF suppression is a proposed mechanism of action, one must assume increased noradrenaline stimulation or increased sensitivity to noradrenaline in the tumor. Studies testing these assumptions in vitro and in vivo on IH models are needed to begin to gain some insight into how propranolol exerts its effects on IH. In our laboratory, we examined the effect of propranolol on VEGF-A production in hemangioma-derived stem cells and endothelial cells, but found no evidence that proproanolol affected VEGF-A under basal cell culture conditions (S Greenberger et al., unpubl.).
Down-Regulation of Other Proangiogenic Cytokines
Angiogenesis is a process in which a coordinated set of events leads to the formation of new blood vessels by sprouting from pre-existing vessels. Among those events is the proteolysis of components of the extracellular matrix allowing the endothelial cells to migrate and sprout (Aznavoorian et al. 1993; Bergers et al. 2000). Accordingly, matrix metalloproteinases (MMPs), secreted both by tumor cells and by tumor-associated macrophages (TAMs) have been shown to be critical for angiogenesis (Bergers et al. 2000; Fang et al. 2000; Huang et al. 2002). In a vascular tumor model, using endothelioma cells, a synthetic inhibitor of MMPs inhibited in vivo tumor growth (Taraboletti et al. 1995). Noradrenaline increases the expression of MMP-2 and MMP-9 by tumor cells and by macrophages, and these effects could be inhibited by the beta-blocker, propranolol (Lutgendorf et al. 2003; Guo et al. 2009). Propranolol has also been reported to block the up-regulation of MMP-7 in a gastric tumor (Shi et al. 2010). In brain vascular endothelial cells, propranolol blocked the induction of MMP-9 by PMA (a phorbol ester that causes an extremely wide range of effects in cells (Blumberg 1981; Annabi et al. 2009). Thus, it is possible that propranolol’s effect is mediated, at least in part, by MMPs regulation.
In addition to MMP down-regulation, propranolol might inhibit hemangiogenesis by the control of another proangiogenic cytokine, IL-6. Recent reports have shown IL-6 is up-regulated by catecholamines, both in the transcriptional and in the translational levels, and that the up-regulation is blocked by propranolol. In-depth analysis of this signaling pathway also suggests a cAMP and Src kinase-dependent mechanism (Nilsson et al. 2007). It is of interest that both corticosteroids and propranolol suppress VEGF-A, MMPs, and IL-6. This overlap in activity may provide a clue to the roles of these proteins in the pathogenesis of IH.
Effects on Differentiation
IH is a tumor in which new blood vessels form, presumably de novo, from immature progenitor (stem) cells. Thus, it is possible that propranolol prevents the hemangioma stem cells from differentiating into endothelial cells or pericytes (Frieden and Drolet 2009). Another possibility is that propranolol hastens the differentiation of the progenitor cells into adipocytes, which would in effect convert the tumor stage from “proliferating” to “involuting.” Although support for this hypothesis has not yet been provided by experimentation with in vitro or in vivo models of IH, effects have been reported on mesenchymal stem cells. For example, noradrenaline has been shown to push mesenchymal progenitors within white adipose tissue toward a brown adipocytic phenotype (Vegiopoulos et al. 2010). Also, an effect of noradrenaline on mesenchymal stem cell adipogenesis has been shown in vitro (Li et al. 2010).
CONCLUDING REMARKS
In recent years, our understanding of the pathological vasculgenic process leading to IH proliferation has expanded with the identification of hemangioma stem cells and the corticosteroid-sensitive targets. The emergence of propranolol as a new therapeutic option that is both safe and effective provides a crucial alternative therapy for disfiguring, and at times, life-endangering hemangiomas. A major challenge for the future years will be to elucidate the mechanism of action of prpranolol on proliferating IH. Insights gained from such studies will enable the development of target-specific drugs, not only for IH but also for malignant vascular tumors and tumor angiogenesis.
Footnotes
Editors: Michael Klagsbrun and Patricia D’Amore
Additional Perspectives on Angiogenesis available at www.perspectivesinmedicine.org
REFERENCES
- Adcock IM, Caramori G, Ito K 2006. New insights into the molecular mechanisms of corticosteroids actions. Curr Drug Targets 7: 649–660 [DOI] [PubMed] [Google Scholar]
- Amir J, Metzker A, Krikler R, Reisner SH 1986. Strawberry hemangioma in preterm infants. Pediatric Dermatol 3: 331–332 [DOI] [PubMed] [Google Scholar]
- Annabi B, Lachambre MP, Plouffe K, Moumdjian R, Beliveau R 2009. Propranolol adrenergic blockade inhibits human brain endothelial cells tubulogenesis and matrix metalloproteinase-9 secretion. Pharmacol Res 60: 438–445 [DOI] [PubMed] [Google Scholar]
- Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M 1995. Immunosuppression by glucocorticoids: Inhibition of NF-κ B activity through induction of I κ B synthesis. Science 270: 286–290 [DOI] [PubMed] [Google Scholar]
- Aznavoorian S, Murphy AN, Stetler-Stevenson WG, Liotta LA 1993. Molecular aspects of tumor cell invasion and metastasis. Cancer 71: 1368–1383 [DOI] [PubMed] [Google Scholar]
- Bandi N, Kompella UB 2001. Budesonide reduces vascular endothelial growth factor secretion and expression in airway (Calu-1) and alveolar (A549) epithelial cells. Eur J Pharmacol 425: 109–116 [DOI] [PubMed] [Google Scholar]
- Benovic JL 2002. Novel β2-adrenergic receptor signaling pathways. J Allergy Clin Immunol 110: S229–S235 [DOI] [PubMed] [Google Scholar]
- Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, Tanzawa K, Thorpe P, Itohara S, Werb Z, et al. 2000. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2: 737–744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bever AT, Hisaw FL, Velardo JT 1956. Inhibitory action of desoxycorticosterone acetate, cortisone acetate, and testosterone on uterine growth induced by estradiol-17β. Endocrinology 59: 165–169 [DOI] [PubMed] [Google Scholar]
- Blumberg PM 1981. In vitro studies on the mode of action of the phorbol esters, potent tumor promoters, part 2. Crit Rev Toxicol 8: 199–234 [DOI] [PubMed] [Google Scholar]
- Boscolo E, Bischoff J 2009. Vasculogenesis in infantile hemangioma. Angiogenesis 12: 197–207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boye E, Yu Y, Paranya G, Mulliken JB, Olsen BR, Bischoff J 2001. Clonality and altered behavior of endothelial cells from hemangiomas. J Clin Invest 107: 745–752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caldenhoven E, Liden J, Wissink S, Van de Stolpe A, Raaijmakers J, Koenderman L, Okret S, Gustafsson JA, Van der Saag PT 1995. Negative cross-talk between RelA and the glucocorticoid receptor: A possible mechanism for the antiinflammatory action of glucocorticoids. Mol Endocrinol 9: 401–412 [DOI] [PubMed] [Google Scholar]
- Chang J, Most D, Bresnick S, Mehrara B, Steinbrech DS, Reinisch J, Longaker MT, Turk AE 1999. Proliferative hemangiomas: Analysis of cytokine gene expression and angiogenesis. Plast Reconstr Surg 103: 1–9 [DOI] [PubMed] [Google Scholar]
- Chang LC, Haggstrom AN, Drolet BA, Baselga E, Chamlin SL, Garzon MC, Horii KA, Lucky AW, Mancini AJ, Metry DW, et al. 2008. Growth characteristics of infantile hemangiomas: Implications for management. Pediatrics 122: 360–367 [DOI] [PubMed] [Google Scholar]
- Cheng JF, Gole GA, Sullivan TJ 2010. Propranolol in the management of periorbital infantile haemangioma. Clin Experiment Ophthalmol 38: 547–553 [DOI] [PubMed] [Google Scholar]
- Cohen SR, Wang CI 1972. Steroid treatment of hemangioma of the head and neck in children. Ann Otol Rhinol Laryngol 81: 584–590 [DOI] [PubMed] [Google Scholar]
- Dadras SS, North PE, Bertoncini J, Mihm MC, Detmar M 2004. Infantile hemangiomas are arrested in an early developmental vascular differentiation state. Mod Pathol 17: 1068–1079 [DOI] [PubMed] [Google Scholar]
- Dalman FC, Bresnick EH, Patel PD, Perdew GH, Watson SJ Jr, Pratt WB 1989. Direct evidence that the glucocorticoid receptor binds to hsp90 at or near the termination of receptor translation in vitro. J Biol Chem 264: 19815–19821 [PubMed] [Google Scholar]
- Das A, Mantena SR, Kannan A, Evans DB, Bagchi MK, Bagchi IC 2009. De novo synthesis of estrogen in pregnant uterus is critical for stromal decidualization and angiogenesis. Proc Natl Acad Sci 106: 12542–12547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dosanjh A, Chang J, Bresnick S, Zhou L, Reinisch J, Longaker M, Karasek M 2000. In vitro characteristics of neonatal hemangioma endothelial cells: Similarities and differences between normal neonatal and fetal endothelial cells. J Cutan Pathol 27: 441–450 [DOI] [PubMed] [Google Scholar]
- Doshi BS, Kulkarni RD, Dattani KK, Anand MP 1984. Effect of labetalol and propranolol on human cutaneous vasoconstrictor response to adrenaline. Int J Clin Pharmacol Res 4: 25–28 [PubMed] [Google Scholar]
- Enjolras O, Mulliken JB 1993. The current management of vascular birthmarks. Pediatr Dermatol 10: 311–313 [DOI] [PubMed] [Google Scholar]
- Fang J, Shing Y, Wiederschain D, Yan L, Butterfield C, Jackson G, Harper J, Tamvakopoulos G, Moses MA 2000. Matrix metalloproteinase-2 is required for the switch to the angiogenic phenotype in a tumor model. Proc Natl Acad Sci 97: 3884–3889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fredriksson JM, Lindquist JM, Bronnikov GE, Nedergaard J 2000. Norepinephrine induces vascular endothelial growth factor gene expression in brown adipocytes through a β-adrenoreceptor/cAMP/protein kinase A pathway involving Src but independently of Erk1/2. J Bioll Chem 275: 13802–13811 [DOI] [PubMed] [Google Scholar]
- Frieden IJ, Drolet BA 2009. Propranolol for infantile hemangiomas: Promise, peril, pathogenesis. Pediatr Dermatol 26: 642–644 [DOI] [PubMed] [Google Scholar]
- Frieden IJ, Haggstrom AN, Drolet BA, Mancini AJ, Friedlander SF, Boon L, Chamlin SL, Baselga E, Garzon MC, Nopper AJ, et al. 2005. Infantile hemangiomas: Current knowledge, future directions. In Proceedings of a research workshop on infantile hemangiomas, April 7–9, 2005, Bethesda, Maryland: Pediatr Dermatol 22: 383–406 [DOI] [PubMed] [Google Scholar]
- Gong H, Jarzynka MJ, Cole TJ, Lee JH, Wada T, Zhang B, Gao J, Song WC, DeFranco DB, Cheng SY, et al. 2008. Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res 68: 7386–7393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenberger S, Adini I, Boscolo E, Mulliken JB, Bischoff J 2010a. Targeting NF-κB in infantile hemangioma-derived stem cells reduces VEGF-A expression. Angiogenesis 13: 327–335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenberger S, Boscolo E, Adini I, Mulliken JB, Bischoff J 2010b. Corticosteroid suppression of VEGF-A in infantile hemangioma-derived stem cells. New Engl J Med 362: 1005–1013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo K, Ma Q, Wang L, Hu H, Li J, Zhang D, Zhang M 2009. Norepinephrine-induced invasion by pancreatic cancer cells is inhibited by propranolol. Oncol Rep 22: 825–830 [DOI] [PubMed] [Google Scholar]
- Haggstrom AN, Drolet BA, Baselga E, Chamlin SL, Garzon MC, Horii KA, Lucky AW, Mancini AJ, Metry DW, Newell B, et al. 2007. Prospective study of infantile hemangiomas: Demographic, prenatal, and perinatal characteristics. J Pediatr 150: 291–294 [DOI] [PubMed] [Google Scholar]
- Hasan Q, Tan ST, Xu B, Davis PF 2003. Effects of five commonly used glucocorticoids on haemangioma in vitro. Clin Exp Pharmacol Physiol 30: 140–144 [DOI] [PubMed] [Google Scholar]
- Huang S, Van Arsdall M, Tedjarati S, McCarty M, Wu W, Langley R, Fidler IJ 2002. Contributions of stromal metalloproteinase-9 to angiogenesis and growth of human ovarian carcinoma in mice. J Natl Cancer Inst 94: 1134–1142 [DOI] [PubMed] [Google Scholar]
- Isik FF, Rand RP, Gruss JS, Benjamin D, Alpers CE 1996. Monocyte chemoattractant protein-1 mRNA expression in hemangiomas and vascular malformations. J Surg Res 61: 71–76 [DOI] [PubMed] [Google Scholar]
- Iwata J, Sonobe H, Furihata M, Ido E, Ohtsuki Y 1996. High frequency of apoptosis in infantile capillary haemangioma. J Pathol 179: 403–408 [DOI] [PubMed] [Google Scholar]
- Jackson R 1998. The natural history of strawberry naevi. J Cutan Med Surg 2: 187–189 [DOI] [PubMed] [Google Scholar]
- Jensen K, Skagen K, Henriksen O, Kjoller E 1983. Local blood flow regulation in normotensive man during prolonged treatment with propranolol. Eur J Clin Pharmacol 24: 723–726 [DOI] [PubMed] [Google Scholar]
- Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, Lin SC, Heyman RA, Rose DW, Glass CK, et al. 1996. A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors. Cell 85: 403–414 [DOI] [PubMed] [Google Scholar]
- Khan ZA, Boscolo E, Picard A, Psutka S, Melero-Martin JM, Bartch TC, Mulliken JB, Bischoff J 2008. Multipotential stem cells recapitulate human infantile hemangioma in immunodeficient mice. J Clin Invest 118: 2592–2599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Lee JM, Park JS, Jo SA, Kim YO, Kim CW, Jo I 2008. Dexamethasone coordinately regulates angiopoietin-1 and VEGF: A mechanism of glucocorticoid-induced stabilization of blood-brain barrier. Biochem Biophys Res Commun 372: 243–248 [DOI] [PubMed] [Google Scholar]
- Kisseleva T, Song L, Vorontchikhina M, Feirt N, Kitajewski J, Schindler C 2006. NF-κB regulation of endothelial cell function during LPS-induced toxemia and cancer. J Clin Invest 116: 2955–2963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinman ME, Greives MR, Churgin SS, Blechman KM, Chang EI, Ceradini DJ, Tepper OM, Gurtner GC 2007. Hypoxia-induced mediators of stem/progenitor cell trafficking are increased in children with hemangioma. Arterioscler Thromb Vasc Biol 27: 2664–2670 [DOI] [PubMed] [Google Scholar]
- Kompella UB, Bandi N, Ayalasomayajula SP 2003. Subconjunctival nano- and microparticles sustain retinal delivery of budesonide, a corticosteroid capable of inhibiting VEGF expression. Invest Ophthalmol Vis Sci 44: 1192–1201 [DOI] [PubMed] [Google Scholar]
- Leaute-Labreze C, Dumas de la Roque E, Hubiche T, Boralevi F, Thambo JB, Taieb A 2008. Propranolol for severe hemangiomas of infancy. New Engl J Med 358: 2649–2651 [DOI] [PubMed] [Google Scholar]
- Lee SK, Kim HJ, Na SY, Kim TS, Choi HS, Im SY, Lee JW 1998. Steroid receptor coactivator-1 coactivates activating protein-1-mediated transactivations through interaction with the c-Jun and c-Fos subunits. J Biol Chem 273: 16651–16654 [DOI] [PubMed] [Google Scholar]
- Li Q, Yu Y, Bischoff J, Mulliken JB, Olsen BR 2003. Differential expression of CD146 in tissues and endothelial cells derived from infantile haemangioma and normal human skin. J Pathol 201: 296–302 [DOI] [PubMed] [Google Scholar]
- Li H, Fong C, Chen Y, Cai G, Yang M 2010. β-Adrenergic signals regulate adipogenesis of mouse mesenchymal stem cells via cAMP/PKA pathway. Mol Cell Endocrinol 323: 201–207 [DOI] [PubMed] [Google Scholar]
- Liu W, Zhang S, Hu T, Jiang X, Hu X, Feng J 1999. Sex hormone receptor of hemangioma and vascular malformation in children. Zhonghua Wai Ke Za Zhi 37: 295–297 [PubMed] [Google Scholar]
- Lutgendorf SK, Cole S, Costanzo E, Bradley S, Coffin J, Jabbari S, Rainwater K, Ritchie JM, Yang M, Sood AK 2003. Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines. Clin Cancer Res 9: 4514–4521 [PubMed] [Google Scholar]
- Maguiness SM, Frieden IJ 2010. Current management of infantile hemangiomas. Semin Cutan Med Surg 29: 106–114 [DOI] [PubMed] [Google Scholar]
- Masuda H, Kalka C, Takahashi T, Yoshida M, Wada M, Kobori M, Itoh R, Iwaguro H, Eguchi M, Iwami Y, et al. 2007. Estrogen-mediated endothelial progenitor cell biology and kinetics for physiological postnatal vasculogenesis. Circ Res 101: 598–606 [DOI] [PubMed] [Google Scholar]
- Mazereeuw-Hautier J, Hoeger PH, Benlahrech S, Ammour A, Broue P, Vial J, Ohanessian G, Leaute-Labreze C, Labenne M, Vabres P, et al. 2010. Efficacy of propranolol in hepatic infantile hemangiomas with diffuse neonatal hemangiomatosis. J Pediatr 157: 340–342 [DOI] [PubMed] [Google Scholar]
- McSorley PD, Warren DJ 1978. Effects of propranolol and metoprolol on the peripheral circulation. Brit Med J 2: 1598–1600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nies AS, Evans GH, Shand DG 1973. Regional hemodynamic effects of β-adrenergic blockade with propranolol in the unanesthetized primate. Am Heart J 85: 97–102 [DOI] [PubMed] [Google Scholar]
- Nilsson MB, Armaiz-Pena G, Takahashi R, Lin YG, Trevino J, Li Y, Jennings N, Arevalo J, Lutgendorf SK, Gallick GE, et al. 2007. Stress hormones regulate interleukin-6 expression by human ovarian carcinoma cells through a Src-dependent mechanism. J Biol Chem 282: 29919–29926 [DOI] [PubMed] [Google Scholar]
- Norman AW, Mizwicki MT, Norman DP 2004. Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov 3: 27–41 [DOI] [PubMed] [Google Scholar]
- Park SY, Kang JH, Jeong KJ, Lee J, Han JW, Choi WS, Kim YK, Kang J, Park CG, Lee HY 2011. Norepinephrine induces VEGF expression and angiogenesis by a hypoxia-inducible factor-1α protein-dependent mechanism. Int J Cancer 128: 2306–2316 [DOI] [PubMed] [Google Scholar]
- Pelaia G, Vatrella A, Cuda G, Maselli R, Marsico SA 2003. Molecular mechanisms of corticosteroid actions in chronic inflammatory airway diseases. Life Sci 72: 1549–1561 [DOI] [PubMed] [Google Scholar]
- Phuc Le P, Friedman JR, Schug J, Brestelli JE, Parker JB, Bochkis IM, Kaestner KH 2005. Glucocorticoid receptor-dependent gene regulatory networks. PLoS Genet 1: e16 10.1371/journal.pgen.0010016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qu Z, Liebler JM, Powers MR, Galey T, Ahmadi P, Huang XN, Ansel JC, Butterfield JH, Planck SR, Rosenbaum JT 1995. Mast cells are a major source of basic fibroblast growth factor in chronic inflammation and cutaneous hemangioma. Am J Pathol 147: 564–573 [PMC free article] [PubMed] [Google Scholar]
- Razon MJ, Kraling BM, Mulliken JB, Bischoff J 1998. Increased apoptosis coincides with onset of involution in infantile hemangioma. Microcirculation 5: 189–195 [PubMed] [Google Scholar]
- Rhen T, Grissom S, Afshari C, Cidlowski JA 2003. Dexamethasone blocks the rapid biological effects of 17β-estradiol in the rat uterus without antagonizing its global genomic actions. Faseb J 17: 1849–1870 [DOI] [PubMed] [Google Scholar]
- Rosbe KW, Suh KY, Meyer AK, Maguiness SM, Frieden IJ 2010. Propranolol in the management of airway infantile hemangiomas. Arch Otolaryngol Head Neck Surg 136: 658–665 [DOI] [PubMed] [Google Scholar]
- Sahlin L 1995. Dexamethasone attenuates the estradiol-induced increase of IGF-I mRNA in the rat uterus. J Steroid Biochem Mol Biol 55: 9–15 [DOI] [PubMed] [Google Scholar]
- Sans V, de la Roque ED, Berge J, Grenier N, Boralevi F, Mazereeuw-Hautier J, Lipsker D, Dupuis E, Ezzedine K, Vergnes P, et al. 2009. Propranolol for severe infantile hemangiomas: Follow-up report. Pediatrics 124: e423–e431 [DOI] [PubMed] [Google Scholar]
- Sasaki GH, Pang CY, Wittliff JL 1984. Pathogenesis and treatment of infant skin strawberry hemangiomas: Clinical and in vitro studies of hormonal effects. Plast Reconstr Surg 73: 359–370 [DOI] [PubMed] [Google Scholar]
- Schaaf MJ, Cidlowski JA 2002. Molecular mechanisms of glucocorticoid action and resistance. J Steroid Biochem Mol Biol 83: 37–48 [DOI] [PubMed] [Google Scholar]
- Schoneveld OJ, Gaemers IC, Lamers WH 2004. Mechanisms of glucocorticoid signalling. Biochim Biophys Acta 1680: 114–128 [DOI] [PubMed] [Google Scholar]
- Shi M, Liu D, Duan H, Han C, Wei B, Qian L, Chen C, Guo L, Hu M, Yu M, et al. Catecholamine up-regulates MMP-7 expression by activating AP-1 and STAT3 in gastric cancer. Molecular Cancer 9: 269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shmelkov SV, Meeus S, Moussazadeh N, Kermani P, Rashbaum WK, Rabbany SY, Hanson MA, Lane WJ, St Clair R, Walsh KA, et al. 2005. Cytokine preconditioning promotes codifferentiation of human fetal liver CD133+ stem cells into angiomyogenic tissue. Circulation 111: 1175–1183 [DOI] [PubMed] [Google Scholar]
- Takahashi K, Mulliken JB, Kozakewich HP, Rogers RA, Folkman J, Ezekowitz RA 1994. Cellular markers that distinguish the phases of hemangioma during infancy and childhood. J Clin Invest 93: 2357–2364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan ST, Wallis RA, He Y, Davis PF 2004. Mast cells and hemangioma. Plast Reconstr Surg 113: 999–1011 [DOI] [PubMed] [Google Scholar]
- Tao Y, Williams-Skipp C, Scheinman RI 2001. Mapping of glucocorticoid receptor DNA binding domain surfaces contributing to transrepression of NF-κ B and induction of apoptosis. J Biol Chem 276: 2329–2332 [DOI] [PubMed] [Google Scholar]
- Taraboletti G, Garofalo A, Belotti D, Drudis T, Borsotti P, Scanziani E, Brown PD, Giavazzi R 1995. Inhibition of angiogenesis and murine hemangioma growth by batimastat, a synthetic inhibitor of matrix metalloproteinases. J Natl Cancer Inst 87: 293–298 [DOI] [PubMed] [Google Scholar]
- Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M, et al. 2006. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat Med 12: 939–944 [DOI] [PubMed] [Google Scholar]
- van Batenburg MF, Li H, Polman JA, Lachize S, Datson NA, Bussemaker HJ, Meijer OC 2010. Paired hormone response elements predict caveolin-1 as a glucocorticoid target gene. PLoS One 5 e8839 10.1371/journal.pone.0008839 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vandenburg MJ, Conlon C, Ledingham JM 1981. A comparison of the effects of propranolol and oxprenolol on forearm blood flow and skin temperature. Br J Clin Pharmacol 11: 485–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vegiopoulos A, Muller-Decker K, Strzoda D, Schmitt I, Chichelnitskiy E, Ostertag A, Berriel Diaz M, Rozman J, Hrabe de Angelis M, Nusing RM, et al. 2010. COX-2 controls energy homeostasis in mice by de novo recruitment of brown adipocytes. Science 328: 1158–1161 [DOI] [PubMed] [Google Scholar]
- Young R, Glennon RA 2009. S(-)Propranolol as a discriminative stimulus and its comparison to the stimulus effects of cocaine in rats. Psychopharmacology 203: 369–382 [DOI] [PubMed] [Google Scholar]
- Yu Y, Varughese J, Brown LF, Mulliken JB, Bischoff J 2001. Increased Tie2 expression, enhanced response to angiopoietin-1, and dysregulated angiopoietin-2 expression in hemangioma-derived endothelial cells. Am J Pathol 159: 2271–2280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Y, Flint AF, Mulliken JB, Wu JK, Bischoff J 2004. Endothelial progenitor cells in infantile hemangioma. Blood 103: 1373–1375 [DOI] [PubMed] [Google Scholar]
- Zarem HA, Edgerton MT 1967. Induced resolution of cavernous hemangiomas following prednisolone therapy. Plast Reconstr Surg 39: 76–83 [DOI] [PubMed] [Google Scholar]
- Zhang L, Lin X, Wang W, Zhuang X, Dong J, Qi Z, Hu Q 2005. Circulating level of vascular endothelial growth factor in differentiating hemangioma from vascular malformation patients. Plast Reconstr Surg 116: 200–204 [DOI] [PubMed] [Google Scholar]
- Zhou F, Bouillard B, Pharaboz-Joly MO, Andre J 1989. Non-classical antiestrogenic actions of dexamethasone in variant MCF-7 human breast cancer cells in culture. Molec Cell Endocrinol 66: 189–197 [DOI] [PubMed] [Google Scholar]