transforming growth factor- superfamily in meningiomas ...paper.uscip.us/ijn/ijn.2015.1001.pdf ·...

20
Columbia International Publishing International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20 doi:10.7726/ijn.2015.1001 Review ______________________________________________________________________________________________________________________________ *Corresponding e-mail: [email protected] 1 Dept. of Pathology and Laboratory Medicine, Univ. of Rochester Medical Center, 601 Elmwood Ave. Box 626, Rochester, NY 14642 1 Transforming Growth Factor- Superfamily in Meningiomas: Targets for Novel Therapy in Meningiomas? Mahlon D. Johnson 1* MD, PhD Received 6 January 2015; Published online 18 April 2015 © Mahlon D. Johnson 2015. Published with open access at www.uscip.us Abstract Meningiomas are central nervous system tumors with a high recurrence rate. In the absence of effective chemotherapies, inoperable, recurrent and metastatic tumors remain a therapeutic challenge. Members of the TGF- super-family play an integral role in the development, progression and metastases of many malignancies yet may be underappreciated participants in the biology of meningiomas. This paper reviews the common abnormalities in TGF- or BMP signaling in neoplasias with a focus on meningiomas. Sites of potential targeting for new chemotherapies are also noted. Keywords: Meningioma; TGF- receptors; TGF-; BMP; SMAD 1. Introduction Meningiomas represent a common primary brain tumor in adults (Perry et al., 1997; Mawrin and Perry, 2010). Although the majority are benign, approximately 20% are World Health Organization (WHO) grade II with a recurrence rate approaching 40% at 5 years after gross total resection (Perry et al. 1997; Mawrin and Perry, 2010). Metastases also occur in grade II tumors with an underestimated frequency (Surov et al., 2013, Forest et al., 2014). Anaplastic, WHO grade III meningiomas, approximately 3-5% of tumors, have an even higher recurrence rate and metastatic potential. Moreover, the mortality rate of anaplastic meningiomas is 20% at 2 years (Forest et al., 2014; Yamaguchi et al., 2014). Thus, while surgical resection is possible for many WHO grade I and II tumors, growth at inaccessible sites complicates management of some. Stereotactic radiotherapy is a treatment option and effective in many cases (Sheehan et al., 2010, Komotar et al., 2012, Starke et al., 2012) but the high rate of recurrence and resistance to radiation over time complicates this

Upload: truongphuc

Post on 12-Mar-2019

216 views

Category:

Documents


0 download

TRANSCRIPT

Columbia International Publishing International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20 doi:10.7726/ijn.2015.1001

Review

______________________________________________________________________________________________________________________________ *Corresponding e-mail: [email protected] 1 Dept. of Pathology and Laboratory Medicine, Univ. of Rochester Medical Center, 601 Elmwood Ave. Box

626, Rochester, NY 14642

1

Transforming Growth Factor- Superfamily in Meningiomas: Targets for Novel Therapy in

Meningiomas?

Mahlon D. Johnson1* MD, PhD

Received 6 January 2015; Published online 18 April 2015 © Mahlon D. Johnson 2015. Published with open access at www.uscip.us

Abstract Meningiomas are central nervous system tumors with a high recurrence rate. In the absence of effective chemotherapies, inoperable, recurrent and metastatic tumors remain a therapeutic challenge. Members of the TGF- super-family play an integral role in the development, progression and metastases of many malignancies yet may be underappreciated participants in the biology of meningiomas. This paper reviews the common abnormalities in TGF- or BMP signaling in neoplasias with a focus on meningiomas. Sites of potential targeting for new chemotherapies are also noted.

Keywords: Meningioma; TGF- receptors; TGF-; BMP; SMAD

1. Introduction Meningiomas represent a common primary brain tumor in adults (Perry et al., 1997; Mawrin and Perry, 2010). Although the majority are benign, approximately 20% are World Health Organization (WHO) grade II with a recurrence rate approaching 40% at 5 years after gross total resection (Perry et al. 1997; Mawrin and Perry, 2010). Metastases also occur in grade II tumors with an underestimated frequency (Surov et al., 2013, Forest et al., 2014). Anaplastic, WHO grade III meningiomas, approximately 3-5% of tumors, have an even higher recurrence rate and metastatic potential. Moreover, the mortality rate of anaplastic meningiomas is 20% at 2 years (Forest et al., 2014; Yamaguchi et al., 2014). Thus, while surgical resection is possible for many WHO grade I and II tumors, growth at inaccessible sites complicates management of some. Stereotactic radiotherapy is a treatment option and effective in many cases (Sheehan et al., 2010, Komotar et al., 2012, Starke et al., 2012) but the high rate of recurrence and resistance to radiation over time complicates this

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

2

management and underscores the need for an effective chemotherapy (Wen et al., 2010; Chamberlain, 2012). The absence of viable chemotherapies has prompted the search for novel therapies targeting growth regulatory cytokines. Of these, members of transforming growth factor beta (TGF-B) super-family may be particularly relevant.

2. TGF-

2a. TGF- synthesis and latency

Fig. 1. TGF- and BMP signaling in meningiomas.

Multiple sites in these receptors and signaling pathways have been identified in malignancies including meningiomas. The transforming growth factor-s (TGF-) represent an important family of numerous peptides including the TGFB1, 2, 3 and bone morphogenetic proteins (BMPs). TGF-s are synthesized as prohormones. Pro- TGF-1, 2 or 3, 75 kDa homodimers, are cleaved in the Golgi apparatus to form 25 kDa homodimers (Barnard et al., 1990; Shi and Massague, 2003). The mature homodimers associate with latency associated proteins to form latency complexes (Barnard et al., 1990; Rifkin, 2005). These are further processed in the cells endoplasmic reticulum where disulfide bonds are

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

3

formed between the latentcy binding protein and TGF- to form larger latent complexes. The larger complexes are secreted to the extracellular matrix where they anchor to cell membrane components such as fibronectin and remain inactive (Taiple et al., 1994; Isogagai et al., 2003). Anchored latent TGF- is cleaved by several proteinases such as thrombospondin and metalloproteinase MMP2, integrins and changes in pH to form the active TGF- monomer which, in turn, binds by sulfides to become functional TGF- (Barcellos-Hoff and Dix, 1990; Schultz-Cherry S and Murphy-Ulrich JE, 1993; Munger, J.S. et al., 1999; Jenkins, 2008).

In terms of growth regulatory effects on normal cells, TGF- is largely inhibitory. In fact, TGF-s inhibit proliferation of nearly all normal epithelial cells, mesenchymal cells and cells from some low grade neoplasias (Piel and Roberts, 2001; Millet and Zhang, 2007; Massague, 2008; Massague, 2012). TGF- effects are largely transduced through binding to the TGF- type II receptor (TGF-RII) that then complexes with the TGF- type I receptor phosphorylating and activating its serine/threonine kinase (TGF-RI) (Massague, 1992). TGF-RIII i.e. betaglycan, is a membrane glycoprotein which facilitates TGF- binding to the TGF-RII (Massague, 1992). The activated TGF-RI receptor phosphorylates a number of membrane/cytoplasmic proteins which transduce growth regulatory signals to the nucleus (Massague, 1992; Massague, 2012) (Figure 1). 2b. TGF- signaling The main signal effectors for TGF- are the SMADs 2 and 3 (Zhang et al., 1996; Zhang and Derynk, 1999). Activation of the type I receptor cytoplasmic domain phosphorylates the carboxyterminal domain of SMAD 2 and SMAD 3 resulting in their heteromerization with SMAD 4 in the cytoplasm. The activated SMAD 2, 3 and 4 complex translocates to the nucleus where they interact with and stabilize transcriptional complexes of numerous transcription factors (Zhang et al., 1996; Zhang and Derynk, 1999; Massague, 2012) (Figure 1). Under some circumstances, TGF- also utilizes other signaling pathways including the RAF-MAPK/Erk kinase-1(MEK-1)-mitogen-activated protein kinase (MAPK) pathway (Yan et al., 1994; Kawabata et al., 1995; Mucasi et al., 1996; Frey and Mulder, 1997; Lewis et al., 1998; Mulder, 2000; Javelaud and Mauviel, 2005). This may be by direct TGFBR1 activation of Ras then downstream RAF, MEK-1 and p44/42MAPK (Mulder, 2000; Javelaud and Mauviel, 2005). This activation may be part of a tightly regulated feedback loop in some tumors such as prostatic carcinoma cells where constitutively activated MAPK induces TGFB synthesis (Yu et al., 2010). Nonetheless, at higher levels TGF- may attenuate p44/42 MAPK activation by recruiting and activating phosphatase PP2a which dephosphorylates MAPK (Yu et al., 2010). TGFBs also signal via the phosphoinositide 3 kinase (PI3K)-protein kinase B/Akt - p70S6K (Kawabata et al., 1995; Lewis et al., 1998; Higaki and Shimmkado, 1999; Vicancano and Sawyers, 2002) and p38-JNK pathways (Frey and Mulder 1997, Ravanti et al 1999). 2c. TGF- expression and meningiomas Altered expression of, or sensitivity to TGF- has been identified in several malignancies where reduction of TGF-s inhibitory effects are thought to contribute to their development. The role of TGF-s in meningioma development is likely specific to degree of differentiation and target cells. TGF-ß1, 2 and 3 as well as TGF-ß receptors I, II and III are expressed in human leptomeninges and in varying levels depending on the grade in meningiomas (Johnson et al., 1992a; Johnson et al.,

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

4

1992b; Johnson et al., 2004; Johnson et al., 2011). Both precursor leptomeningeal cells and meningioma cells synthesize and release all three (presumably latent) TGF- isoforms in vitro (Johnson et al. 1992a; Johnson et al. 1992b; Johnson et al. 2004; Johnson et al. 2011). At least in vitro, TGF-1 reduces basal proliferation in half of WHO grade I meningioma cell cultures (Johnson et al., 1992a; Johnson et al., 2004) and attenuates epidermal growth factor induced proliferation in 80% of meningiomas (Johnson et al., 1992a). Thus, paracrine TGF-1 effects from leptomeningeal cells may tonically suppress growth of human leptomeningeal and WHO grade I meningioma cells (Johnson et al. 1992, Johnson et al., 2004). Autocrine TGF-1 may also restrain grade I meningioma growth (Johnson et al., 1992a; Johnson et al., 2004). In WHO grade I meningioma cells this inhibitory TGF- signaling is associated with TGF-1 activation/phosphorylation of SMAD 2/3 (Johnson et al., 2004). In contrast, the p44/42 MAPK pathway does not appear activated by TGF-1 in WHO grade I meningiomas (Johnson et al., 2004). In contrast, the p44/42 MAPK pathway, which is mitogenic to meningioma cells (Johnson et al., 2001; Johnson et al., 2002; Mawrin et al., 2005) is activated by TGF-1that stimulates phosphorylation of p44/42 MAPK in some meningioma cell cultures from grade II meningiomas. Thus, p44/42 MAPK may transduce some TGF- effects in higher grade meningioma cells with low, but still partially functional receptors (Johnson et al., 2011). As meningiomas progress and TGF- RII levels decline, non-SMAD, non-inhibitory pathways may participate in meningioma biology. 2d. Loss of TGF- inhibition and meningiomas A reduction in sensitivity to TGF-’s peptide’s growth inhibition may facilitate development of several malignancies (Glick et al., 1994 Fan et al., 1989). For example, disruption of TGF- promotes progression in squamous cell carcinoma (Glick et al., 1994). This scenario is also seen in astrocytes and low grade astrocytoma cells that secrete TGF--1 and 2 and are inhibited by exogenous TGF- (Jennings et al., 1991). However, anaplastic astrocytoma cells become progressively resistant to the growth inhibitory effects of TGF- as they evolve into malignant gliomas (Jennings et al., 1991). Similarly, intestinal epithelium and benign adenoma cells are also inhibited by TGF- but become progressively resistant to TGF- as adenoma cells dedifferentiate into carcinomas (Manning et al., 1991; Principe et al., 2014). Paracrine TGF-1 from leptomeningeal cells may tonically suppress growth of human leptomeningeal and WHO grade I meningioma cells (Johnson et al., 1992a, 1992b, 2004). Autocrine TGF-1 may also restrain grade I meningioma proliferation (Johnson et al., 2004). In WHO grade I meningioma cells this inhibitory TGF- signaling is associated with TGF-1 activation/phosphorylation of SMAD 2/3 (Johnson et al., 2004). In contrast, TGF- inhibition is not detected in most WHO grade II and III meningiomas (Johnson et al., 2011). Thus, loss of sensitivity to TGF-'s inhibitory effects may also occur in higher grade meningiomas as well as other tumors. 2e. TGF- immunosupression in meningiomas Immunomodulation is a central role of TGF-1 which has potent immunosupressive effects on natural killer and cytotoxic T cells (Shull et al., 1999). In many neoplsms, TGF- supression of the body's immunosurveillance is thought to contribute to the progression and metastases of malignancies (Massague, 2008). Inflammatory infiltrates have been described at the invading edge

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

5

of meningiomas (Grund et al., 2009). Nonetheless, the role of the immune system in limiting the meningioma invasion and metastases has not been extensively studied. Future studies may clarify the importance of inflammatory and TGF- immunomodulatory effects on meningioma invasion, spread and recurrence. 2f. Alterations in TGF- receptors in neoplasms including meningiomas Loss of TGF-Rs has been described in numerous other malignancies. Missense or frameshift mutations in the TGF-RI has been demonstrated in breast, ovarian, esophageal and head and neck carcinomas (Ohue et al. 1996; Chen et al., 1998; Kretzschmar 2000; Knobloch et al., 2001; Wang et al., 2007; Xu and Pasche et al., 2007; Jin et al. 2008; Massague, 2008; Massague, 2012; Chen et al., 2011). Epigenetic decreased expression in TGF-RI has been found in gastric carcinoma (Massague, 2008). Inactivating mutations in TGF-RI, often in tumors with microsatellite instability, has been found in breast, ovarian, esophageal and head and neck carcinomas (Chen et al., 1999 and Chen et al., 2001). Mutations in TGF-RII have been found in pulmonary, biliary, gastric, colonic and ovarian carcinomas (Ohue et al., 1996; Chen et al., 1998; Kretzschmar, 2000; Matsushita et al., 2005; Wang et al., 2007; Xu and Pasche, 2007; Jin et al., 2008; Chen et al., 2011). Loss or down regulation of the TGF-RIII may be even more common in malignancies having been identified in prostatic and non-small cell carcinoma, neuroblastomas, ovarian, endometrial and renal cancers (Iolascon et al. 2000; Copland et al. 2003; Florio et al., 2005; Hempel et al., 2007; Sharfi et al., 2007; Turley et. al, 2007; Finger et al. 2008; Bilandzic et al. 2009;). Loss of TGF-RIIIs may also permit progression of breast cancer (Dong et al., 2007). The role of altered TGF-R expression in the pathogenesis of meningiomas is not fully understood. TGF-RII protein is reduced or undetectable in the majority of grade II and III meningiomas. TGF-RIII mRNA is significantly reduced in WHO grade III compared to lower grade meningiomas. In addition, TGF-1 inhibition of leptomeningeal and WHO grade I meningioma cell growth, (Johnson et al., 1992a; Johnson et al., 2004; Johnson et al., 2011), is not detected in nearly all WHO grade II and the grade III tumor cells. Recent, comparative genomic hybridization has also identified a 2.3 fold reduction in TGF-RIII gene copy in “high proliferative” meningiomas (Carvalho et al., 2007). Thus, because TGF- effects are initiated by binding to the TGF- type II and III receptors, a genetic or epigenetic reduction in the concentration of receptors likely results in attenuated TGF- growth inhibition. This may permit less restrained growth of higher grade meningiomas. 2g. SMAD 7 inhibition of TGF- signaling in meningiomas SMAD 7 is a natural inhibitor of TGF- signaling and part of complex feedback loops regulating TGF- effects (Kleef et al., 1999; Massague, 2008; Massague, 2012). It has been hypothesized that SMAD 7 might increase in higher grade meningiomas as another mechanism of escape from TGFBs inhibitory effects. Overexpression of SMAD 7, attenuating TGF-s inhibitory effects, has been found in endometrial and thyroid carcinomas (Nakano et al., 1997; Cerutti et al., 2003; Dowdy et al., 2005). EGF also increases SMAD 7 which is inhibitory to SMAD 2/3 signaling (Dowdy, 2006). Nonetheless, SMAD 7 expression is not increased or significantly different between meningioma grades (Johnson et al., 2011). Moreover, a recent comparative genomic hybridization analysis has found a 3 fold reduction rather than increase in SMAD 7 gene expression in high grade compared to low grade meningiomas (Carvalho et al., 2007).

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

6

2h. Alterations in TGF- signaling in meningiomas Loss of sensitivity to the inhibitory effects of TGF- in meningiomas may also be downstream from alterations in receptor activation, SMAD or MAPK signaling. Mutations in Smad 4 have been identified in pancreatic carcinoma and murine adenocarcinoma of the colon (Luttges et al., 2001; Xu et al., 2007). Reduced Smad signaling has also been associated with development of carcinomas of the head and neck and metastatic potential (Xie et al., 2003a; Xie et al., 2003b). LMO4, which influences TGF- signaling by interactions with SMAD signaling, represent a downstream site where inhibitory TGF- signaling may be altered (Lu et al., 2006). Moreover, this appears to be in concert with mitogenic signals from other growth factors. For example, activation of mitogenic EGF receptor tyrosine kinases (present on meningioma cells) activate MAPK/Erk kinase which hyperphosphorylates SMAD 2 and 3 preventing their movement into the nucleus and TGF- signaling (Principe et al., 2014). TGF-1 also influences cell proliferation by alternately reducing p44/42 MAPK (erk) phosphorylation at high concentrations while increasing phosphorylation/activation at lower concentrations particularly in higher grade tumors (Principe et al., 2014; Zhang et al., 2014). In our studies, TGF-1 increased p44/42MAPK phosphorylation in the majority of higher grade i.e. grade II meningiomas (Johnson et al., 2011). This regulation appears to be by influencing the activation of protein phosphatase 2a (PP2A) which can alter p-44/42MAPK phosphorylation status (Lu et al., 2006). Thus, early in the development of meningiomas, other factors may slowly override any tonic inhibitory effects on meningioma cell proliferation. Higher grade, particularly anaplastic meningiomas have the highest recurrence rate and the least response to any current therapy. Our findings suggest that restoring TGF- inhibitory signaling pathways may be an important component to the development of effective chemotherapies for meningiomas. 2i. Cerebrospinal fluid and TGF- signaling in meningiomas TGF-1 is present in human cerebrospinal fluid at concentrations that may activate TGF- receptors (Vawter et al., 1996; Johnson et al., 1992b; Johnson et al., 2004). Nonetheless, its' influence on meningioma cell growth appears complex. Cerebrospinal fluid, from patients without neurological disease, is a potent mitogen for meningioma cells of all grades in vitro (Johnson et al., 2012). In this context, TGF-1 in cerebrospinal fluid appears to limit grade I meningioma cell proliferation in vitro. For example, adding TGF-1 neutralizing antibodies to cerebrospinal fluid potentiates the cerebrospinal fluid’s mitogenic effects in some meningioma cells (Johnson et al. 2004). Cerebrospinal fluid effects on TGF- isoforms and TGF- receptors have not been reported. However, screening of a limited number of meningiomas in vitro found that cerebrospinal fluid (from patients without neurological disease of inflammation) had no effect on the TGF- precursor protein levels in cells from 5 human meningiomas (Johnson et al., unpublished). In these same cells, cerebrospinal fluid had no effect on protein levels of the TGF- antagonist, SMAD 7. However, intriguingly, exogenous platelet derived growth factor-BB (PDGF-BB), which is also in cerebrospinal fluid and released by meningioma cells, increased SMAD 7 levels in meningioma cells (Johnson et al., unpublished). This raises the possibility that cerebrospinal fluid’s mitogenic effects

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

7

on meningioma cells may, in part, be via induction of SMAD 7 expression which could counteract TGF-'s inhibitory autocrine or paracrine effects. 2j. TGF- and Merlin in Meningiomas Expression of TGF-, TGF-Rs, SMAD 7and phosphorylation of SMAD 3 do not correlate with the presence or absence of Merlin protein, the tumor suppressor protein encoded by the NF2 gene (Gusella et al., 1999; Perry et al., 2004, Riemenschneider et al., 2006) in any grade of meningiomas. Consequently, Merlin doesn’t appear to regulate TGF-, TGF-Rs or SMAD 3 phosphorylation.

3. BMPs The bone morphogenic proteins (BMP) include 20 dimers that are part of the larger transforming growth factor beta (TGF-ß) superfamily (Chen et al., 2004; Herpin and Cunningham, 2007). While BMPs were originally identified as proteins inducing bone formation, it is now recognized that they influence multiple functions including cell proliferation, apoptosis, angiogenesis, differentiation and extra cellular matrix formation (Chen et al., 2004; Herpin and Cunningham, 2007). BMP4 is synthesized as a large precursor peptide which is subsequently cleaved to form the active carboxyl terminus protein and secreted as a dimer (Chen et al., 2004; Herpin and Cunningham, 2007). BMPs signal through binding and coupling of two BMP type II and type I receptor serine/threonine kinases forming a heterotetracomplex. The activated BMPRII receptors transphosphorylate the type I receptors which, in turn, phosphorylate intracellular substrates including Smad I, p38 MAPK and other signaling proteins (Chen et al., 2004; Herpin and Cunningham, 2007). 3a. BMP signaling BMP4 influences cellular function by binding cooperatively to the BMPRI and II, forming a heterotetraplex of 2 BMPRIIs and 2 BMPRIs. The constitutively active type II receptors transphosphorylate and activate the type I receptor serine/threonine kinases (Miyazono et al., 2005; Herpin and Cunningham, 2007). Major signaling is thru phosphorylation of Smad 1, 5 and 8 which form a heterocomplex with Smad4 and enter the nucleus where they directly regulate or couple with Co-Smads to regulate transcription factors influencing target gene transcription (Miyazono et al., 2005; Herpin and Cunningham, 2007). BMP4 also signals by activation of non-SMAD pathways such as the p 38 MAPK, p44/42 MAPK and JNK pathways (Herpin and Miyazono et al., 2005; Jeffrey et al., 2005; Beck et al., 2007; Cunningham, 2007). 3b. BMP effects on leptomeningeal and other central nervous system cells Evaluation of the effects of BMPs in the leptomeninges has focused on BMP4 and BMP7, the main BMPs isolated there. BMP4 and 7 are inhibitory to many normal cell types (Kallionemi, 2012). The role of BMP4 and BMP7 in regulating leptomeningeal cell proliferation appears complex. BMP4, BMP7, BMP R1a and BMPRIIa are expressed in human fetal and adult leptomeningeal cells (Johnson et al., unpublished data). However, levels of these BMPs may be low as neither BMP4 nor BMP7 can be detected in media conditioned by fetal leptomeningeal cells. Similarly, BMPR2 and BMP4 have been detected in the mouse leptomeninges (Ikeda et al., 1996). Exogenous BMP4 has no effect on

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

8

basal leptomeningeal cell proliferation but inhibits some fetal leptomeningeal cell proliferation in PDGF-BB stimulated cells in vitro. In contrast, BMP7 stimulates some fetal leptomeningeal cell proliferation in vitro (Johnson et al., unpublished data). Paracrine effects of BMP 4 or 7 released form the pia, arachnoid and dura also appear to have major effects on overlying and subjacent tissues. In the brain, BMP4 and 7 inhibit growth and promote differentiation, at least in neuronal and astrocytic lineages (Mabie et al., 1999; Dorsky et al., 2000; Yabe et al., 2002; Gomes et al., 2003; Segklia et al., 2012; Choe et al., 2012; Choe et al., 2013). With regard development of the overlying calvarium, several studies have shown that BMP4 promotes osteoblast and chondrocyte differentiation (Nishimura et al., 2008; Shen et al., 2009; Chen et al., 2012; Nishimura et al., 2012). BMPs also influence angiogenesis in many tissues, in part, by influencing VEGF and VEGF receptor expression. BMP4 reduces VEGF expression in the second and third trimester human fetal leptomeninges (Johnson et al. unpublished observations). Similarly, BMP9 reduces VEGF expression in endothelial cells (Shao et al., 2009). In contrast, BMP4 stimulates VEGFA expression in in osteoblast-derived endothelial cells (Deckers et al., 2009). Thus, BMP4 regulation of VEGF appears cell and tissue specific. BMP4 regulation of angiogenesis in developing tissues and during neoplastic transformation appears to be via activation of SMAD1/5/8 and downstream activation of VEGF family and its receptors (Lowery and de Caestecker, 2010; Farnsworth et al., 2011). Regarding the leptomeninges, BMP effects on VEGF receptors are also isoform dependent. BMP4 has no effect on leptomeningeal VEGFR expression (Johnson et al. unpublished observations). However in endothelial cells, BMP4 increases VEGFR (Suzuki et al., 2008). In contrast, BMP7 increases VEGFR expression s in fetal leptomeninges and this is potentiated PDGFs effects on stimulation (Johnson et al. unpublished observations). To our knowledge, this has not been previously described but may reflect another important role for BMP7 in leptomeningeal development. 3c. BMP expression in meningiomas BMP4 expression has been demonstrated in meningiomas (Johnson et al., 2009) and BMP4 mRNA has been reported in 6 of 7 uncharacterized meningiomas of unknown subtype and grade (Hirota et al., 1995). In WHO grade I tumors, expression is detected in meningothelial, transitional and fibrous meningiomas and not restricted to a specific subtype. BMP4 has also been detected in media conditioned by WHO grade I and II meningiomas (Johnson et al., 2009). In higher grade meningiomas, expression was detected in only fibrous and transitional (Hirota et al., 1995). PCR analysis of low and high grade meningiomas has also found reduced median BMP4, Smad7, Smad 9 and TGF-ß R III expression in grade III compared with grade I meningiomas (Carvalho et al., 2007). Thus, reduced BMP4 gene expression may also be a feature of some higher grade meningiomas possibly contributing to the pathogenesis of these meningiomas. 3d. BMP regulation of proliferation in neoplasia including meningiomas BMPs appear to have more complex effects on cell proliferation and progression of many neoplasms. BMP4 is inhibitory to myeloma, pulmonary adenocarcinoma, breast, gastric and

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

9

pancreatic cancer cells (Hjertner, O. et al. 2001, Buckley, S. et al. 2004; Ketolainen, J.M. et al. 2010, Shirai, Y.T. et al. 2011; Virtanen, S. et al. 2011 ). However, BMP4 has also been implicated in promotion of invasion and/or metastases of many tumors such as breast, colon, and pancreatic carcinoma cells (Deng, H. et al. 2007; Kallioniemi, A. 2012). Of particular note, BMP4 gene variants increasing BMP4 have also been associated with increased risk of developing cancer of the colon and rectum (Houlston, R.S. et al. 2008; Lubbe, S.J. et al. 2012; Slattery, M.L. et al. 2012 ). This complexity also applies to cell proliferation in central nervous system and pituitary tumors. BMPs have been either inhibitory or stimulatory in central nervous system-associated tissues. BMP4 inhibits glioma stem cell proliferation (Zhou et al., 2011; Kallionemi, 2012) but stimulates pituitary adenoma cells and inhibits primitive neuroectodermal cell apoptosis (Intoska, et al., 1999; Paez-Pereda et al., 2003,). BMP4 stimulation of other select mesodermal and epithelial cells has also been described (Yang et al., 2007, Montesano et al., 2008). In meningiomas, BMP4 potentiates DNA/cell proliferation when added to fetal bovine serum. This raises the possibility that BMP4 may potentiate proliferative effects of other growth factors released by meningioma cells or present in cerebrospinal fluid (Johnson et al., 2009). Meningioma cell release of BMP4 raises the possibility of complex interactions between this cytokine, other members of the TGF-ß superfamily (Johnson et al., 2005) and other growth regulatory cytokines secreted by meningioma cells or in the cerebrospinal fluid.

4. Targeting TGF- and BMP Signaling Alterations in Meningiomas

Disrupting alterations in TGF- or BMP signaling in malignancies depends on the site and nature of the defect. To date, direct therapeutic options are limited, in part, due to toxicities associated with disrupting TGF- signaling. Nonetheless, in many malignancies where TGF- switches from inhibitory effects to promotion of tumor progression, TGF- signaling may is being blocked small molecule inhibtors of the TGF- type I receptor (Dituri et al., 2013). Preliminary studies suggest LY 2157229 is effective in blocking TGF- effects. At least in initial human toxicity studies, when carefully dosed, LY 2157229 may have limited cardiac toxicity (Rodon, J et al., 2014). Because TGF- induces retinoic acid which may mediate some signaling thru retinoic acid receptors, targeting the RA signaling pathways may bypass defects in TGF- R III loss in progressively malignant meningiomas (Xu and Kopp, 2012). The role of BMP4 and 7 in treatment of meningiomas is likely to be multifaceted. One potential target might be the disfiguring hyperostosis of the skull overlying sites of meningioma growth. Hyperostosis is a common morbidity associated with meningiomas especially in the calvarium (Goyal et al. 2012) and may require extensive bone resection ( Bikmaz K et al. 2007). A similar reaction has been demonstrated in prostate carcinoma cells that stimulate bone formation via secretion of BMP4. Treatment with monoclonal antibodies to BMP4 and small molecule inhibitors to BMP4 blocked this osteoblastic reaction (Lee et al., 2012). This study raises the possibility that local administration of anti-BMP4 may suppress hyperostosis around meningiomas as well.

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

10

5. Conclusions Loss of TGF- and BMP inhibition of cell proliferation may be central to the development of meningiomas. Alterations in TGF- and BMP signaling pathways may also influence progression, invasion metastasis and immunosurveillance against the development of meningioma cells. Clarification of the multiple sites where TGF- superfamily members affect meningioma biology may facilitate development of novel therapies against these frequently recurrent tumors.

References

Bakin, A.V., Tomlinson, A.K., Bhowmick, N. A., Moses, H.L., Arteaga, C.L. (2000). Phosphatidylinositol 3-kinase

function is required for transforming growth factor B-mediated epithelial to mesenchymal transition

and cell migration. J Biol Chem, 275, 36803-36810.

http://dx.doi.org/10.1074/jbc.M005912200

Barcellos-Hoff, M.H., Dix, T.A. (1990). Redox-mediated activation of latent transforming growth factor-beta 1.

Mol Endocinol, 10, 1077-83.

Barnard, J.A., Lyons, R.M., Moses, H.L. (1990). The cell biology of transforming growth factor B. Biochim

Biophys Acta, 1032, 79-87.

Beck, S.E., Jung, B.H., Del Rosario, E. Gomez, L., Carethers, J.M. (2007). BMP-induced growth suppression in

colon cancer cells is mediated by p21WAF1 stabilization and modulated by RAS/ERK. Cell Signal, 19,

1465-72.

http://dx.doi.org/10.1016/j.cellsig.2007.01.017

Bilandzic, S., Chu, P.G., Farnworth, C. , Harrison C., Nicholis, P., Wang, Y., Escalona, R.M., Fuller, P.J., Findlay, J.K.,

Stenvers, K.L. (2009). Loss of betaglycan contributes to the malignant properties of human granulosa

tumor cells. Mol Enedocrinol, 23, 539-548.

http://dx.doi.org/10.1210/me.2008-0300

Bikmaz, K., Mrak, R., Al-Mefty, O. (2007). Management of bone invasive, hyperostotic sphenoid wing

meningiomas. J Neurosurg, 10, 905-12.

http://dx.doi.org/10.3171/JNS-07/11/0905

Buckley, S., Shi, W. Driscoll, B. Ferrario, A. Anderson, K. Warburton, D. (2004). BMP4 signaling induces

senescence and modulates the oncogenic phenotype of A549 lung adenocarcinoma cells. Am J Physiol

Lung Cell Mol Physiol, 286, L81-6.

http://dx.doi.org/10.1152/ajplung.00160.2003

Carvalho, L.H., Smirnov, I., Baia, G.S., Modrusan, Z., Smith, J.S., Jun, P., Costello, J.F., McDermott, M.W.,

Vandenberg, S.R., Lal, A. (2007). Molecular signatures define two main classes of meningiomas. Mol

Cancer, 6, 64-78.

http://dx.doi.org/10.1186/1476-4598-6-64

Cerutti J.M., Ebina KN, Matsuo S.E., Martins, L., Maciel, R.M., Kimura, E.T. (2003). Expression of Smad4 and

Smad7 in human thyroid follicular carcinoma cell lines. J Endocrinol 26, 516-521.

http://dx.doi.org/10.1007/BF03345213

Chamberlain, M.C. (2012). The role of chemotherapy and targeted therapy in treatment of intracranial

meningioma. Current Opin Oncol, 24, 666-671.

http://dx.doi.org/10.1097/CCO.0b013e328356364d

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

11

Chen G, Deng C, Li Y-P. (2012). TGF-B and BMP signaling in osteoblast differentiation and bone formation. Int

J Biol Sci, 8, 272-288.

http://dx.doi.org/10.7150/ijbs.2929

Chen T., Carter, D., Garrique-Antar, L., Reiss, M. (1998). Transforming growth factor B type I receptor kinase

mutant associated with metastatic breast cancer. Cancer Res, 58, 4805-10.

Chen, T., Triplett, J., Dehner, B., Hurst, B. Colligan, B. Pemberton, J. Graff, J.R. Cater, J.H. (2001). Transforming

growth factor B type I receptor gene is frequently mutated in ovarian carcinomas. Cancer Res, 61, 4679-

8229.

Chen, T., Carter, D., Garrigue-Antar, L, Reis, M. (1999). Transforming growth factor β type receptor kinase

mutant associated with metastatic breast cancer. Cancer Res, 58, 4805-10.

Chen, D., Zhao, M., Mundy, G.R. 2004. Bone morphogenic proteins. Growth Factors 2004, 22, 233-41

http://dx.doi.org/10.1080/08977190412331279890

Choe, Y., Siegenthaler, J.A., Pleasure, S.J. (2012). A cascade or morphogenic signaling initiated by the meninges

controls corpus callosum formation. Neuron, 73, 698-712.

http://dx.doi.org/10.1016/j.neuron.2011.11.036

Choe, Y., Kozlova, A., Graf, D., Pleasure, S.J. (2013). Bone morphogenic protein signaling is a major

determinant of dentate development. J Neuroci, 33, 6766-6755.

http://dx.doi.org/10.1523/JNEUROSCI.0128-13.2013

Copland, J.A., Luxon, A., Ajani, T. maity, T., Campagnaro, E., Guo, H., LeGrand, S.N., Tanboli, P., Wood, C.G.

(2003). Genomic profiling identifies alterations in TGF Beta signaling through loss of TGFBeta receptor

receptor espression in human renal cell carcinogenesis and progression. Oncogene, 22, 8053-8062.

http://dx.doi.org/10.1038/sj.onc.1206835

Deckers, M.M., van Bezooijen, R.L., van der Hors,t G., Van Der Bent, C., Papapoulos, S.E. et al. (2009). Bone

morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth

factor A. Endocrinology, 143, 1545-1553.

http://dx.doi.org/10.1210/endo.143.4.8719

Deng, H. Ravikumar, T.S. Yang, W.L. (2009). Overexpression of bone morphogenetic protein 4 enhances the

invasiveness of Smad4-deficient human colorectal cancer cells. Cancer Lett, 281, 220-231

http://dx.doi.org/10.1016/j.canlet.2009.02.046

Dituri F, Mazzocca A, Peidro FJ, Pappicco P, Fabergat I, DeSantis, T., Paradiso, A., Sabba, C., Gainnelli, G. (2013).

Differential inhibition of the TGF-beta signaling pthway in HCC cells using the small molecule inhibitor

LY2157299 and D10 monoclonal antibody against the TGF-beta receptor type II. Plos ONE, 8:e67109.

http://dx.doi.org/10.1371/journal.pone.0067109

Dong, M., How, T., Kirkbride, K.C. Gordon, K..J., Lee, J.D., Hempel, N., Kelly, P., Moeller, B.J., Marks, J.R., Blobe,

G.C. (2007). The type III TGF-beta receptor suppresses breast cancer progression. J Clin Invest, 17, 206-

17.

http://dx.doi.org/10.1172/JCI29293

Dorsky, R.I., Moon, T.R., Raible, D.W. (2000). Enviornmental signals and cell fate specification in premigratory

neural crest. Bioessays, 22, 708-7716.

http://dx.doi.org/10.1002/1521-1878(200008)22:8<708::AID-BIES4>3.0.CO;2-N

Dowdy, S.C., Mariani, A., Reinholz, M.M., Keeney, G.L., Spelsberg, T.C., Podtarz, K.C., Janknecht, R., (2005).

Overexpression of the TGF-beta antagonist Smad 7 in endometrial cancer. Gynecol Oncol, 96, 368-373.

http://dx.doi.org/10.1016/j.ygyno.2004.10.006

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

12

Fan, D.S., Chakrabarty, C., Seid, C.W., Bell, H. Schackert, K., Morikawa, K., Fidler, I.J.(1989). Clonal stimulation

or inhibition of human colon carcinomas and human renal carcinomas mediated by transforming

growth factor-beta 1. Cancer Commun, 1, 117-125.

Farnsworth, R.H., Karnezis, T., Shayan, R., Matsumoto, M., Nowell, C.J., Achen, M.G. Stacher, S.A. (2011). A role

for bone morphogenetic protein-4 in lymph node vascular remodeling and primary tumor growth.

Cancer Res, 71, 6547-6557.

http://dx.doi.org/10.1158/0008-5472.CAN-11-0200

Finger, E.C., Turley, R.S., Dong, M., How, T., Fields, T.A., Blobe, G.C. (2008). TGFBeta RIII suppresses non-small

cell lung cancer invasiveness and tumorigenicity. Carcinogenesis, 29, 528-535

http://dx.doi.org/10.1093/carcin/bgm289

Frey, R.S., Mulder, K.M. (1997). Involvement of extracellular signal-regulated kinase 2 and stress-activated

protein kinase/Jun N-terminal kinase activation by transforming growth factor B in negative growth

control of breast cancer cells. Cancer Res, 57, 628-633.

Florio, P., Ciarmela, F.M., Reis, P., Toti,P., Galleri, L., Santopetro, R., Tiso, E., Tosi, P., Petraglia, F. (2005). Inhibin

alpha subunit and the inhibin co-receptor betaglycan are downregulated in endometrial carcinoma. Eur

J Endocrinol, 152, 277-284

http://dx.doi.org/10.1530/eje.1.01849

Forest, F. Berremilla S-A, Gyenes, C., Ginguene C, Kassir R, Sulalman A, Pasquier, B., Porcheron, J., Peoc'h M.

(2014). Metastatic meningiomas: an unusual clinical and pathological diagnosis with highly variable

outcome. J Neurooncol, 120, 411-421.

http://dx.doi.org/10.1007/s11060-014-1567-2

Glick, A.B., Lee, M.M., Darwiche, N., Kulkarni, A.B., Karlsson, S., Yuspa, S.H. (1994). Targeted deletion of the

TGF-beta gene causes rapid progression to squamous cell carcinoma. Genes Dev, 8, 2429-2440.

http://dx.doi.org/10.1101/gad.8.20.2429

Gomes, W.A., Mehler, M.F., Kessler, J.A. (2003). Transgenic overexpression of BMP4 increases astroglial and

decreases oligodendroglial lineage commitment. Dev Biol, 255, 164-177

http://dx.doi.org/10.1016/S0012-1606(02)00037-4

Goval, N., Kakkar, A. Sarkar, C., Agrawal, D. (2012). Does bony hyperostosis in intracranial meningiomas

signify tumor invasion? a radio-pathologic study. Neurol India, 60: 50-4.

http://dx.doi.org/10.4103/0028-3886.93589

Grund, S., Schittenhelm, J., Rosenert. F. Tatagiba, M., Mawrin, C., Kim, Y.J., Bornemann, A. (2009). The

micorglial/macrophagic response at the tumour-brain border of invasive meningiomas. Neuropathology

and Applied Neurobiol, 35, 82-88.

http://dx.doi.org/10.1111/j.1365-2990.2008.00960.x

Gusella, J.F., Ramesh, V., MacCollin, M., Jacoby, L.B. (1999). Merlin: the neurofibromatosis 2 tumor suppressor.

Biochem Biophys Acta, 1423, M29-36.

http://dx.doi.org/10.1016/S0304-419X(99)00005-0

Hempel, N., How, T., Dong, S.K. Murohy SK, fields TA, Blobe GC. (2007). Loss of betaglycan expression in

ovarian cancer: role in motility and invasion. Cancer, 6, 5231-5238.

http://dx.doi.org/10.1158/0008-5472.CAN-07-0035

Herpin, A., Cunningham, C. (2007). Cross-talk between the bone morphogenic protein pathway and other

major signaling pathways results in tightly regulated cell-specific outcomes. FEBS Journal, 274, 2977-

2985.

http://dx.doi.org/10.1111/j.1742-4658.2007.05840.x

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

13

Hiertner, O., Hjorth-Hansen, H., Borset, M., Seidel, C., Waage, A., Sundan, A. (2001). Bone morphogenetic

protein-4 inhibits proliferation and induces apoptosis of multiple myeloma cells. Blood, 97, 516-22.

http://dx.doi.org/10.1182/blood.V97.2.516

Higaki, M., Shimokado, K. (1999). PI3K. Arterioscler Thromb Vasc Biol, 19, 2127-2132.

http://dx.doi.org/10.1161/01.ATV.19.9.2127

Hirota, S., Yoshikazu, N., Yoshimine, T. Kohri, K., Nomura, S., Taneda, M., Hayakawa, T., Kitamura, Y., (1995).

Expression of bone-related protein messenger RNA in human meningiomas: Possible involvement of

osteopontin in development of psammoma bodies. J Neuropathol Exp Neurol, 54, 698-703.

http://dx.doi.org/10.1097/00005072-199509000-00012

Houlston, R.S. Webb, E. Broderick, P. et al. (2008). Met-analysis of genome-wide association with data

identifies four new susceptibility loci for colorectal cancer. Nat Genet, 40, 1426-1435.

http://dx.doi.org/10.1038/ng.262

Ikeda, T., Takahashi, H., Suzuki, A., et al. (1996). Cloning of rat type I receptor cDNA for bone morphogenic

protein-2 and bone morphogenic protein-4 and the localization compared with that of the ligands. Dev

Dynamics, 206, 319-329.

http://dx.doi.org/10.1002/(SICI)1097-0177(199607)206:3<318::AID-AJA9>3.0.CO;2-L

Intosca, M.R.,McPherson, C.E., Ho, S.Y., Maxwell, G.D. (1999). Bone morphogenetic proteins-2 and-4 attenuate

apoptosisin a cerebellar primitive neuroectodermal tumor cell line. J Neurosci Res, 56, 248-258.

http://dx.doi.org/10.1002/(SICI)1097-4547(19990501)56:3<248::AID-JNR4>3.0.CO;2-U

Iolascon, L., Giodani, A., Borrielo, R., Carborne, R, Izzo, A., Tonini, G.P., Gambini, C., Dell, R.F. (2000). Reduced

expression of transforming growth factor–beta receptor type III in high stage neuroblastomas. Br J

Cancer, 82, 1171-1176.

Isogai. Z., Ono, R.N., Ushiro. S. Keene, D.R., Chen, Y., Mazzieri, R., Charbonneau, N.L., Reinhaedt, D.P., Rifkin, D.B.,

Sakai, L.Y. (2003). Latent transforming growth factor beta-binding protein 1 interacts with fibrillin and

is a microfibril-associated protein J Biol Chem, 278, 2750-2757.

http://dx.doi.org/10.1074/jbc.M209256200

Javelaud, D., Mauviel, A. (2005). Cross talk mechanisms between the mitogen-activated protein kinase

pathways and Smad signaling downstream of TGF-beta: implications for carcinogenesis. Oncogene, 24,

5742-50.

http://dx.doi.org/10.1038/sj.onc.1208928

Jeffrey, T.K., Upton, P.D., Trembath, R.C., Morrell, M.W. (2005). BMP4 inhibits proliferation and promotes

myocyte differentiation of lung fibroblasts via Smad1 and JNK pathways. Am J Physiol Lung Cell Mol

Physiol, 288, L370-8.

http://dx.doi.org/10.1152/ajplung.00242.2004

Jenkins G. (2008). The role of proteases in transforming growth factor-B activation. Int J Biochem, Cell Biol 40,

1068-1078.

http://dx.doi.org/10.1016/j.biocel.2007.11.026

Jennings, M.T., Maciunas, R.J., Carver, R., Bascom, C.C., Juneau, P., Misulis, K., Moses, H.L. (1991). TGF-

TGF-

support of an autocrine hypothesis. Int J Cancer, 4, 129-139

http://dx.doi.org/10.1002/ijc.2910490124

Jin, G., Deng, Y., Miao, R. et al. (2008). TGFB1 and TGFBR2 functional polymorphisms and risk of esophageal

squamous cell carcinoma: a case-control analysis in the chinese population. J Cance Res Clin Oncol, 134,

345-351.

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

14

http://dx.doi.org/10.1007/s00432-007-0290-1

Johnson, M.D., Federspiel, C.F., Gold, L.I, Moses, H.L. (1992a). Transforming growth factor-β and transforming

growth factor-β receptor expression in human meningioma cells. Am J Path, 141, 631-642.

Johnson, M.D., Gold, L.I., and Moses, H.L.(1992b). Evidence for TGF-β expression in human leptomeningeal

cells and TGF-β-like activity in human cerebrospinal fluid. Lab. Invest, 67:360-368.

Johnson, M.D., Woodard, A., Kim, P., Frexes-Steed, M. (2001). Evidence for mitogen associated protein kinase

activation and transduction of mitogenic signals from platelet derived growth factor in human

meningioma cells. J Neurosurg, 94, 303-310.

http://dx.doi.org/10.3171/jns.2001.94.2.0293

Johnson, M.D., Woodard, A., Okediji, E.J., et. al. (2002). Lovastatin is a potent inhibitor of meningioma cell

proliferation: evidence for inhibition of a mitogen associated protein kinase. J Neurooncol, 6, 133-142.

http://dx.doi.org/10.1023/A:1014588214966

Johnson, M.D., Okediji, E., Woodard, A. (2004). Transforming growth factor-

proliferation and signal transduction pathways. J Neurooncol, 66, 9-166.

http://dx.doi.org/10.1023/B:NEON.0000013461.35120.8a

Johnson, M.D., Toms, S. (2005). Mitogenic signal transduction pathways in meningiomas: novel targets for

meningioma chemotherapy? J Neuropathol Exp Neurol, 64, 1029-1036.

http://dx.doi.org/10.1097/01.jnen.0000189834.63951.81

Johnson, M.D., Vito, F., O'Connell, M.J., Pilcher, W. (2009). Bone Morphogenetic Protein-4 and receptors are

expressed in the leptomeninges and meningiomas and signal via MAPK. J Neuropathol Exp Neurol ,

68,1177-1183.

http://dx.doi.org/10.1097/NEN.0b013e3181bc6642

Johnson, M.D., Shaw, A.K., O'Connell, M.J., Sim, F.J., Moses, H.L. (2011). Analysis of transforming growth factor

b receptor expression and signaling in higher grade meningiomas. J Neurooncol. 103, 277-85.

http://dx.doi.org/10.1007/s11060-010-0399-y

Johnson MD, O'Connell M, Facik M, Maurer P, Jahromi B, Webster P. (2012) Cerebrospinal fluid stimulates

leptomeningeal and meningioma cell proliferation and activation of STAT3. J. Neurooncol, 107:121-133

http://dx.doi.org/10.1007/s11060-011-0736-9

Kallionemi, A. (2012). Bone morphoegetic protein 4- a fascinating regulator of cancer behavior. Cancer Gen,

205, 267-287.

http://dx.doi.org/10.1016/j.cancergen.2012.05.009

Kawabata, M., Imanura, T., Miyazono, K., Engel, M.E., Moses, H.L. (1995). Interaction of the transforming

growth factor-B type 1 receptor with farnesyl-protein transferase-a. J Biol Chem, 270, 29628-29631.

http://dx.doi.org/10.1074/jbc.270.50.29628

Ketolainen, J.M., Alarmo, E.L., Tuominen, V.J., Kallionemi, A. (2010). Parallel inhibition of cell growth andf

induction of cell migration and invasion of breast cancer cells by bone morphogenetic protein 4. Breast

Cancer Res Treat, 124, 377-386.

http://dx.doi.org/10.1007/s10549-010-0808-0

Kleef, J. ,Ishiwata, T. Maruyama, H., Friess,H., Truong, P., Buchler M.W. , Falb, D,. Korc, M. (1999). The TGF-Beta

signaling inhibitor enhances tumorigenicity in pancreactic cancer. Oncogene, 18: 5363-5373.

Knobloch, T.J., Lynch, M.A., Song, H. DeGroff, V.L., Casto, B.C., Adams, E.M., Alam, K.Y., Lang, J.C., Schuller, D.E.,

Weghorst, C.M. (2001). Analysis for TGF-B type I receptor for mutations in polymorphisms in head and

neck cancers. Mutation Res, 479, 131-139.

http://dx.doi.org/10.1016/S0027-5107(01)00157-9

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

15

Komotor , R.J. Iorgulescu, J.B. Raper D.M.S, Holland EC, Beal, K.Bilsky, M.H., Brennan C.W., Tabar, V. Sherman,

J.H.Yamada, Y., Gutin, P.H. (2012). The role of radiotherapy following gross total resection of atypicl

meningiomas. J Neurosurg. 117: 679-686.

Kretzschmar, M., (2000). Transforming growth factor-B and breast cancer. Transforming growth factor-

B/SMAD signaling defects and cancer. Breast Cancer Res, 2,107-115.

Lee, Y-C., Cheng C-J, Bilen, M.A., Lu, J-F, Satcher , R.L. Yu-Lee L.Y., Gallick, G.E., Maity S.N., Lin, S.H. (2011).

BMP4 promotes prostate tumor growth in bone through osteogenesis. Cancer Res, 71, 5194-5203.

http://dx.doi.org/10.1158/0008-5472.CAN-10-4374

Lewis, T.S., Shapiro, P.S., Ahn, N.G. (1998). Signal transduction through MAP Kinase cascades. Adv Cancer Res,

74, 49-139.

http://dx.doi.org/10.1016/S0065-230X(08)60765-4

Lowery, J.W. and de Caestecker, M.P. (2010). BMP signaling in vascular development and disease. Cytokine

and Growth Factor Reviews , 21, 287-298. 32.

http://dx.doi.org/10.1016/j.cytogfr.2010.06.001

Lu, Z., Lam, K.S., Wang, N., Xu, X., Cortes, M., Andersen B. (2006). LMO4 can interact with SMAD proteins and

modulate transforming growth factor-beta signaling in epithelial cells. Oncogene, 25, 2920-2930.

http://dx.doi.org/10.1038/sj.onc.1209318

Lubbe, S.J. Pittmari, A.M., Olver, B. , Lloyd, A., Valayakrishnan, L., Naranjo, S., Dobbins, S., Brodernick, P.,

Gomez-Skarmeta, J.L. Houslston, R.S. (2012). The 14q22.2 colorectal cancer variant rs4444235 shows

cis-acting regulation of BMP4. Oncogene, 31, 3777-84.

http://dx.doi.org/10.1038/onc.2011.564

Luttges, J., Galehdari H, Brocker, V. Schwarte-Waldhoff, L. Henne-Bruns, D. Kloppel, G., Schmiegel, W. Hahn,

S.A., 2001. Allelic loss is often the first hit in the biallelic inactivation of DPC4 genes during pancreatic

carcinogenesis. Am J Pathol, 158: 1677-1683.

Mabie, P.C., Mehler, M.F., Kessler, J.A. (1999). Multiple roles of bone morphogenetic signaling in the regulation

of cortical cell number and phenotype. J Neurosci, 19, 7077-7088.

Manning, A.M., Williams, A.C., Game, S.M., Paraskeva, C. (1991). Differential sensitivity of human colonic

adenoma and carcinoma cells to transforming growth factor beta (TGF-Beta): conversion of an adenoma

cell line to a tumorigenic phenotype is accompanied by a reduced response to the inhibitory effects of

TGF-beta. Oncogene :1471-1476

Massaque, L. (1992). Receptors for the TGF- Cell, 69, 1067-1070.

http://dx.doi.org/10.1016/0092-8674(92)90627-O

Massague, J. (2008). TGF-B in cancer. Cell, 134, 215-230.

http://dx.doi.org/10.1016/j.cell.2008.07.001

Massague J. (2012). TGFB signaling in context Nat Rev Mol Cell Biol, 13, 616-630.

Matsushita, M., Matsuraki, K., Date, M., Wantanabe, T., Shibano, K., Nakagawa, T., Yanagitani, S., Amoh, Y.,

Takemoto, H., Ogata, N., Yamoto, S. (1999). Down-regulation of TGF-

cancer: implications for cancer development. Br J Cancer, 80, 194-205.

http://dx.doi.org/10.1038/sj.bjc.6690339

Mawrin, C., Sasse, T., Kirches, E., Kropf S, Schneider ,T. Grimm, C.,Pambor,C., Vorwerk, C. Firsching, R.,

Lendeckel, U., Dietzmann, K. (2005). Different activation of mitogen activated protein kinase and Akt

signaling is associated with aggressive phenotype of human meningiomas. Clin Cancer Res, 11, 4074-

4082.

http://dx.doi.org/10.1158/1078-0432.CCR-04-2550

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

16

Mawrin, C. Perry, A. (2012). Pathological classification and molecular genetis of meingiomas. J Neurooncol, 99,

379-391.

http://dx.doi.org/10.1007/s11060-010-0342-2

Millet, C., Zhang, Y.E., (2007). Roles of Smad 3 in TGF-B signaling during carcinogenesis. Crit Rev Eukaryot

Gene Expr. 17, 281-293.

http://dx.doi.org/10.1615/CritRevEukarGeneExpr.v17.i4.30

Miyazono, K., Kusanagi, K., Inoue, H. (2001). Divergence and convergence of TGF-

Phys , 187, 265-276.

http://dx.doi.org/10.1002/jcp.1080

Miyazono, K., Maeda, S., Imamura, T. (2005). BMP receptor signaling: Transcriptional targets, regulation of

signals and signaling cross-talk. Cytokine and Growth Factor Reviews, 16, 251-263.

http://dx.doi.org/10.1016/j.cytogfr.2005.01.009

Montesano R, Sarkozi R, Schramek H. (2008). Bone morphogenic protein-4 strongly potentiates growth

factor-induced proliferation of mammary epithelial cells. Biochem Biophys Res Commun, 374: 164-168.

Mucsi, I., Skorecki, K.L., Goldberg, H.J. (1996). Extracellular signal-regulated kinase and the small GTP-binding

protein, Rac, contribute to the effects of transforming growth factor-B1 on gene expression. J Biol Chem,

271, 16567-16572.

http://dx.doi.org/10.1074/jbc.271.28.16567

Mulder, K.M. (2000). Role of Ras and Mapks in TGFB signalling. Cytokines Growth Factor Rev, 11,23-3515.

http://dx.doi.org/10.1016/S1359-6101(99)00026-X

Munger JS, Huang X, Kawakatsu, H. Griffiths M.J., DAlton, S.L., Wu, J., Pittel, J.F., Kaminski, N., Garat, C., Matthay,

M.A., Rifkin, D.B., Sheppard, D, (1999). The integrin alpha v beta 6 binds and activates latent TGF beta 1:

a mechanism for regulatingpulmonary inflammation and fibrosis. Cell, 96, 319-328.

http://dx.doi.org/10.1016/S0092-8674(00)80545-0

Nakano, A., Afrakhte, M., Moren, A., Nakayama, T., Christian, J.L., Heuchel, R., Itoh, S., Kawabata, M., Heldin, H.E.,

Heldin, C.H., ten Dijke, P. (1997). Identification of Smad 7 a TGF- e antagonist of TGF-

signaling. Nature, 389, 631-635.

http://dx.doi.org/10.1038/39369

Nishimura, R., Hata, K, Ikeda, F., Ichida, F., Shimoyama, A., Matsubara, T., Wada, M., Amano, K. and Yoneda, T.

(2008). Signal transduction and transcriptional regulation during mesenchymal cell differentiation . J

Bone Miner Metabol, 26, 203-212.

http://dx.doi.org/10.1007/s00774-007-0824-2

Nishimura, R., Hata, K., Matsubara, T., Wakabayshi, M., Yoneda, T. (2012). Regulation of bone and cartilage

development by network between BMP signaling and transcription factors. J Biochem, 151, 247-254.

http://dx.doi.org/10.1093/jb/mvs004

Ohue, M., Tomita, N., Monden, T., Miyoshi, Y., Ohnishi, T., Izawa, H., Kawabata, Y., Sasaki, M., Sekimoto, M.,

Nishisho, I., Shiozaki, H., Monden, M. (1996). Mutations of the transforming growth factor receptor type

II receptor gene and microsatellite instability in gastric cancer. Int J Cancer, 68, 203-6.

http://dx.doi.org/10.1002/(SICI)1097-0215(19961009)68:2<203::AID-IJC11>3.0.CO;2-B

Paez –Pereda, M., Giacomini, D., Refojo, D., Nagashima, A.C., Hopfer, U., Grubler, Y., Chervin, A. Golberg, V.,

Goya, R., Hentges, S.T., Low, M.W. (2003). Involvement of bone morphogenetic protein 4 (BMP-4) in

pituitary prolactinoma pathogenesis through a Smad/estrogen receptor crosstalk. Proc Natl Acad Sci

USA, 100, 1034-1039.

http://dx.doi.org/10.1073/pnas.0237312100

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

17

Perez-Magain E, Campos-Martin Y, Mur P, Fiano C, Ribalta T, Garcia JF, Rey JA. Rodriquez de Lopes, A., Mollejo,

M., Melendez, B. (2012). Genetic alterations associated with progression and recurrence in meningiomas.

J Neuropathol Exp Neurol, 71, 882-893.

http://dx.doi.org/10.1097/NEN.0b013e31826bf704

Perry, A., Stafford, S.L., Scheithauer, B.W., et al.(1997). Meningioma grading: an analysis of histologic

parameters. Am J Surg Pathol, 21, 1455-1465.

http://dx.doi.org/10.1097/00000478-199712000-00008

Perry, A., Gutman, D.H., Reifenberger, G. (2004). Molecular pathogenesis of meningiomas. J Neuroncol,

70,183-202.

http://dx.doi.org/10.1007/s11060-004-2749-0

Piel, E., Roberts, A.B., (2001). Suppressor and oncogenic roles of transforming growth factor-B and its

signaling pathways in tumorigenesis. Adv Cancer Res, 38, 1-49.

Principe, D.R., Doll, J.A., Bauer, J., Jung, B., Munshi, H.G., Bartholin, L., Pasche,B., Lee, C., Grippo, P.J., (2014).

TGFB: Duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst. 106, 369-

388.

http://dx.doi.org/10.1093/jnci/djt369

Ravanti, L., Hakkinen,, l, Larjava, H., Saarialho-Kere, U., Foschi, M., Han, J., Kahari, V.M. (1999). Induction of

collagenase-3 (MMP-13) expression in human skip fibroblasts by three-dimensional collagen is medited

by p38 mitogen-activated protein kinase. J Biol Chem, 274, 37292-37300.

http://dx.doi.org/10.1074/jbc.274.52.37292

Riemenschneider, M.J., Perry, A., Reifenberger, G. (2006). Histological classification and molecular genetics of

meningiomas. Lancet Neurol, 5, 1045-1054.

http://dx.doi.org/10.1016/S1474-4422(06)70625-1

Rifkin, D.B. (2005). Latent transforming growth factor-beta (TGF-beta) binding proteins: orchestrators of

TGF-Beta availability. J Biol Chem, 280, 7409-7412.

http://dx.doi.org/10.1074/jbc.R400029200

Rodon, J., Carducci, M.A., Sepulveda-Sanchez, J.M., Azaro, A., Calvo, E. et al. (2014). First-in-human dose study

of the novel transforming growth factor-B receptor 1 kinase inhibitor LY 2157299 monohydrate in

patients with advanced cancer and glioma. Clin Cancer Res, Epub Nov 25,2104.

Schultz-Cherry, S., Murphy-Ullrich, J.E. (1993). Thrombospondin causes activation of latent transforming

growth factor-beta secreted by endothelila cells by a novel mechanism. J Cell Biol, 122, 923-932.

http://dx.doi.org/10.1083/jcb.122.4.923

Segklia, A., Seuntjens, E., Elkouris, M., Tsalavos, S., Stappers, E. Mitsiadis, T.A., Huylebroek, D., Remboutsika, E.,

Graf, D. (2012). BMP7 regulates the survival, proliferation and neurogenic properties of neural

progenitor cells during corticogenesis in the mouse. PLOS One. 7, 34088.

http://dx.doi.org/10.1371/journal.pone.0034088

Sharifi, N., Hurt, E.M, Kawasaki, B.T., Kawasaki, B.T., Farrar, W.L. (2007). TGFBR3 loss and consequences in

prostate cancer. Prostate, 67, 301-311.

http://dx.doi.org/10.1002/pros.20526

Shao, E.S., Lin, L., Yao, Y., Bostrom, K.I. (2009). Expression of vascular endothelial growth factor is

coordinately regulated by the activin-like kinase receptors 1 and 4 in endothelial cells. Blood, 114, 2197-

2208.

http://dx.doi.org/10.1182/blood-2009-01-199166

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

18

Shen Q, Zhu S, Hu J, Geng N, Zhou S. (2009). Recombinant human bone morphogenetic protein-4 (BMP-4)-

stimulated cell differentiation and bone formation within expanding calvarial sutures in rats. J Craniofac

Surg 20, 1561-5.

http://dx.doi.org/10.1097/SCS.0b013e3181b09cc1

Sheehan, J.P., Williams, B.J., Yen, C.P. (2010). Stereotactic radiosurgery for WHO grade I meningiomas. J

Neurooncol, 99, 407-416.

http://dx.doi.org/10.1007/s11060-010-0363-x

Shi, Y., Massague, J. (2003). Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell, 113,

685-700.

http://dx.doi.org/10.1016/S0092-8674(03)00432-X

Shirai, Y.T. Ehata, S., Yashiro, M., Yanagihara, K., Hirakawa, K., Miyazono, K. (2011). Bone morphogenetic

protein-2 and -4 play tumor suppressive roles in human diffuse-type gastric carcinoma. Am J Pathol, 179,

2920-30.

http://dx.doi.org/10.1016/j.ajpath.2011.08.022

Shull, M.M., Ormsby, I., Kier, A.B., Pawlowski S., Diebold, R.J., Yin, M., Allen, R., Sidman, C., Proetzel, G., Calvin, D.

Et al. (1992). Targeted disrution of the mouse transforming growth factor-beta 1 gene results in

multifocal inflammatory disease. Nature, 359:693-699.

Slattery, M.L. Lundgreen, A., Herrick, J.S., Kadlubar, S., Caan, B.J., Potter, J.D., Wolff, R.K. (2012). Genetic

variation in bone morphogenetic protein and colon and rectal cancer. Int J Cancer, 130, 653-664.

http://dx.doi.org/10.1002/ijc.26047

Starke, R.M., Williams, B.J., Hiles, C., Nguyen, J.H., Elsharkawy, M.H., Sheehan, J.P. (2012). Gamma knife surgery

for skull base meningiomas. J Neurosurg, 116: 588-597, 2012

http://dx.doi.org/10.3171/2011.11.JNS11530

Surov, A., Gottschling, S., Bloz, J., Kornhuber, M., Alfieri, A., Holtzhausen H-J, Abbas, J., Kosling, S., (2013).

Distant metsatases in meningioma: an underestimated problem. J Neurooncol, 112, 323-327.

http://dx.doi.org/10.1007/s11060-013-1074-x

Suzuki, Y., Montagne, K., Nishihara, A., Watabe, T., Miyazono, K. (2008). BMPs promote proliferation and

migration of endothelial cells via stimulation of VEGF-A/VEGFR2 and angiopoietin-1/Tie2 signalling. J

Biochem, 143, 199-206.

http://dx.doi.org/10.1093/jb/mvm215

Taiple, J., Miyazono, K., Heldin, C.H., Keski-Oja, J. (1994). Latent transforming growth factor-beta 1 associates

to fibroblast extracellular matrix via latent TGF-beta binding protein J Cell Biol, 124,171-181.

Turley, R.S., Finger, E.C., Hempel, N., How, T., Fields, T.A., Blobe, G.C. (2007). The type III transforming growth

factor – beta receptor as a novel tumor suppressor gene in prostate cancer. Cancer Res, 67, 1090-1098.

http://dx.doi.org/10.1158/0008-5472.CAN-06-3117

Virtanen, S., Alarmo, E.l., Sandstrom, S., Ampuja, M., Kallionemi, A. 2011. Bone morphogenetic protein -5 and -

5 in pancreatic cancer-novel bidirectional players. Exp Cell Res 317, 2136-46.

Vivanco, I., Sawyers, C.L. (2002). The phosphatidylinositol 3-kinase–AKT pathway in human cancer. Cell

Signal, 14, 381-95.

http://dx.doi.org/10.1038/nrc839

Vawter, M.P., Dillon-Carter, O., Tourtellote, W.W., Carvey, P., Freed, W.J. (1996). TGF-beta 1 and TGF beta 2

concentrations are elevated in Parkinson's disease in ventricular cerebrospinal fluid. Exp Neurol,

142,313-22.

http://dx.doi.org/10.1006/exnr.1996.0200

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

19

Wang, J.C., Su, C.C., Xu, J.B., Chen, L.Z., Hu, X.H., Wang, G.Y., Bao, Y., Huang, Q., Fu, S.B., Li, P., Lu, C.Q., Zhang, R.M.,

Luo, Z.W. (2007). Novel microdeletion in the transforming growth factor B type II receptor gene is

associated with giant and large cell variants of nonsmall cell lung carcinoma. Genes, Chromosomes

Cancer, 26, 192-201.

http://dx.doi.org/10.1002/gcc.20400

Wen , P. Y., Quant, E., Drapatz, J. , Beroukhim, R. Norden, A.D. (2010). Medical therapies for meningiomas. J

Neurooncol, 99, 365-378.

http://dx.doi.org/10.1007/s11060-010-0349-8

Xie, W. , Bharathy, S., Kim, D. Haffty, B.G. Rimm, D.L. Reiss, M. (2003a). Frequent alterations of Smad signaling

in human head and neck squamous cell carcinomas: a tissue microarray analysis. Oncol Res, 14: 61-73.

Xie, W. , S., Rimm, D.L.Lin, Y. Shih W.J., Reiss, M. (2003b). Loss of Smad signaling in human colorectal cancer is

associated with advanced disease and prognosis. Cancer J, 9, 302-312.

Xu,Q., Kopp, J. 2012. Retinoid and TGF-B families: Crosstalk in development, neoplasia, immunity and tissue

repair. Sem Nephrol, 32: 287-294.

Xu, X., Pasche, B, (2007). TGF-B signaling alterations and susceptibility to colorectal cancer. Hum Mol Genet,

16, R14-20.

http://dx.doi.org/10.1093/hmg/ddl486

Yabe, T., Samuels, I., Schwartz, J.P., 2002. Bone morphogenetic proteins BMP-6 and BMP-7 have differential

effects on survival and neurite outgrowth of cerebellar granule cell neurons. J Neuroscience Res, 68,

161-168.

Yamaguchi, S., Terasaka,S., Kobayashi, H, Asaoka, K., Motegi, H., Nishihara, H., Kanno, H., Onimaru, R., Ito, Y.M.,

Shirato, H., Houkin, K. (2014). Prognostic factors for survival in patients with high-grade meningiomas

and recurrence-risk stratification for application of radiotherapy. PLO S, 9, e97108

http://dx.doi.org/10.1371/journal.pone.0097108

Yan, Z., Winawer, S., Friedman, E. (1994). Two different signal transduction pathways can be activated by

-13237.

Yang X, Lee PJ, Long L et al. (2007). BMP induces HO-1 via Smad-independent, p38MAPK-dependent pathway

in pulmonary artery myocytes. Am J Respir Cell Mol Biol, 37, 598-605.

http://dx.doi.org/10.1165/rcmb.2006-0360OC

Yu, N., Kozlowski JM, Park, II, Chen, L., Zhang, Q., Xu, D., Doll, J.A., Crawford, S.E., Brendler, C.B. Lee, C. (2010).

Overxpression of transfroming growth factor beta 1 in malignant prostate cells is partly caused by a

runaway of TGF-beta1auto-induction mediated theough a defective recruitment of protein phosphatase

2a by TGF-beta type I receptor. Urology, 76; 1519-1530.

Zhang, Y., Feng, X-H, Wu, R.Y., Derynk, R. (1996). Receptor associated Mad homologues synergize as effectors

of the TGF- ure, 383, 168-172.

http://dx.doi.org/10.1038/383168a0

Zhang, Y., Derynk, R. (1999). Regulation of Smad signaling by protein associations and signaling crosstalk.

Trend Cell Biol, 9, 274-9.

http://dx.doi.org/10.1016/S0962-8924(99)01579-2

Zhang, Q., Yu, N., Lee, C. (2014). Mysteries of TGF-B paradox in benign and malignant cells. Front Oncol, 4, 94-

99.

http://dx.doi.org/10.3389/fonc.2014.00094

Mahlon D. Johnson / International Journal of Neuropathology (2015) Vol. 3 No. 1 pp. 1-20

20

Zhou Z, Sun L, Wang, Y., Wu, Z., Geng, J., Miu, W., Pu, Y., You, Y., Zhang, Z., Liu, N. (2011). Bone morphogenetic

protein 4 inhibits cell proliferation and induces cell apoptosis in glioma stem cells. Cancer Biother

Radiopharm, 26, 77-83.

http://dx.doi.org/10.1089/cbr.2010.0857