6 Molecular Biology of Meningiomas: Tumorigenesis and Growth



10.1055/b-0034-81185

6 Molecular Biology of Meningiomas: Tumorigenesis and Growth

Ragel Brian T., Jensen Randy L.

Introduction


Meningiomas are generally characterized as slow-growing tumors derived from arachnoid cap cells ( Fig. 6.1 ). Meningiomas are graded as benign (~92% of meningiomas), atypical (6%), or anaplastic/malignant (4%), based on histological characteristics.1 More recently, molecular factors have been found to correlate with tumor grade, and they are the focus of this chapter ( Fig. 6.2 ). Genetic abnormalities are especially important in meningioma tumorigenesis, particularly loss of heterozygosity (LOH) of chromosome 22 and neurofibromatosis 2 (NF2) gene mutations. The role of radiation in meningioma development is also discussed. Furthermore, tumor growth may result from oncogene- or growth factor–mediated growth dysregulation. Studies have implicated various growth factors, including the major mediator of tumor angiogenesis, vascular endothelial growth factor (VEGF). The section on the role of sex hormones in meningioma tumorigenesis highlights hormones associated with tumor growth as well as the association between tumor progression and progesterone receptor loss. Finally, the possible involvement of the inflammatory cascade mediated through cyclooxygenase-2 (COX-2) is explored. Overall, this chapter provides an overview of the current knowledge of meningioma molecular biology with an emphasis on the history, clinical observations, and laboratory models that have made this possible.



Chromosomal Alterations in Meningiomas


Meningiomas were among the first tumors analyzed for genetic abnormalities. This was first performed by using Giemsa staining, but over time more sophisticated methods, including fluorescence in situ hybridization (FISH), comparative genomic hybridization, and spectral karyo-typing, have confirmed and characterized these genetic changes. Chromosome 22 abnormalities, usually in the form of LOH or partial deletion of 22q, are the most frequent abnormalities in all meningioma types ( Fig. 6.3 ).1,2 Meningiomas occurring in the setting of NF2 always exhibit chromosome 22q abnormalities, whereas roughly 50% of meningiomas occurring sporadically exhibit 22q abnormalities. Chromosome 1 abnormalities have been associated with aggressive tumor phenotype.1,3 In general, karyotypic aberrations increase with meningioma tumor grade ( Fig. 6.2 ).3 In addition to 1q loss, chromo-some abnormalities associated with higher-grade meningiomas include 6q, 10p, 10q, 14q, and 18q ( Fig. 6.2 ).1,3



Chromosome 22q:NF2Gene and the Gene Protein Product, Schwannomin/merlin


The link between chromosome 22 abnormalities and meningiomas was first suspected in patients with NF2. The hallmark of this disease is bilateral acoustic schwannomas. Interestingly, meningiomas occur in ~50% of patients with NF2. The NF2 tumor suppressor gene, located on chromosome 22q12.1, and its protein product, schwannomin or merlin ( Figs. 6.3 and 6.4 ), have been identified in cytogenetic and molecular studies.1,2 The NF2 gene codes for the schwannomin/merlin tumor suppressor (TuS) protein (moesin-, ezrin-, radixin-like protein), which is a part of the band 4.1 families of cyto-skeleton-associated proteins ( Fig. 6.4 ; Table 6.1 ).1,2 Possible functions of schwannomin/merlin include roles in cytoskeletal functions (e.g., contact inhibition) and secondary signaling pathways (e.g., Ras) ( Fig. 6.4 ).2,4 Insertions or deletions of this gene produce a nonfunctional merlin protein, resulting in decreased cell adhesion and tumorigenesis.4 Reduced schwannomin/merlin expression has been demonstrated in sporadic meningiomas.5

Fig. 6.1 Factors involved with meningioma tumorigenesis. Arachnoid cap cells (upper inset, cross) are the cell of origin. Known meningioma inciting events and factors associated with tumor growth are listed. (Lower left) Coronal T1-weighted magnetic resonance image with contrast showing parasagittal meningioma with histologic features consistent with meningothelial subtype (lower inset, World Health Organization grade I) with lobules (asterisk) surrounded by thin fibrous septa (plus) and intranuclear halos (arrow). COX-2, cyclooxygenase-2; NF, neurofibromatosis; LOH, loss of heterozygosity; VEGF, vascular endothelial growth factor. Figure modified, with permission from Ragel BT, Jensen RL, Couldwell WT. Inflammatory response and meningioma tumorigenesis and the effect of cyclooxygenase-2 inhibitors. Neurosurg Focus 2007;23(4):E7.
Fig. 6.2 Factors involved with meningioma tumor progression. Loss of chromosome 1p is a decisive step. Immunohistochemically, higher-grade tumors are associated with decreased progesterone receptor (PR) staining and increased MIB-1 nuclear staining. Human telomerase reverse transcriptase (hTERT) activity is also increased in higher-grade meningiomas. LOH, loss of heterozygosity; NF2, neurofibromatosis 2; WHO, World Health Organization.
Fig. 6.3 (A) Representative G-band karyotype of a benign meningioma exhibiting the classic finding of monosomy 22 (i.e., loss of heterozygosity) (arrow). (B) Ideogram of the short (p) and long (q) arms of chromosome 22. Arrows indicate locations of proposed meningioma tumor suppressor genes. Figure reprinted with permission from Glick et al, 1989.23
Fig. 6.4 Schematic of chromosome 22 containing the neurofibromatosis 2 (NF2) gene, which encodes for the tumor suppressor (TuS) protein schwannomin/merlin. Possible functions of the schwannomin/merlin TuS protein include roles in cytoskeletal functions and secondary signaling pathways.









































































Table 6.1 Meningioma Chromosomal Abnormalities and Their Corresponding Genes, Proteins, and Protein Function

Chr


Gene (s)


Protein (s)


Function (s)


Note (s)


22q12.1


NF2


Merlin/schwannomin


Regulation of cell growth and motility


Merlin protein is structurally similar to the protein 4.1 (DAL-1) superfamily


22q12


β-adaptin(BAM22)


BAM22 protein


Intracellular transport of receptor–ligand complexes


Beta-Adaptin-Meningioma-chromosome 22


22q12.3-q13.1


LARGE


LARGE protein


Synthesis of glycoprotein and glycosphingolipid sugar chains


Human-like acetylglucosaminlyltransferase


22q11


MN1


MN1 protein


Transcriptional regulation



22q


INI1 (SMARCB1/hSNF5)


INI1 protein


Transcriptional regulation



1p


der (1)(1qter? 1p11::22q12? 22pter)


Unknown


Unknown


1p13 implicated in radiation-induced meningiomas


9p21


CDKN2A (p16INK4α) P14ARF CDKN2B (p15INK4b)


CDKN2A (p16INK4α) protein P14ARF protein CDKN2B (p15INK4b) protein


Cell-cycle checkpoint proteins



17q22


BRIT1


DEAH-box DNA helicase


BRCA1-dependent DNA repair and checkpoint functions


BRCA interacting protein 1, breast cancer susceptibility gene 1


18p11.3


DAL-1



Cytoskeletal protein


Member of 4.1 protein superfamily


Modified from Ragel and Jensen.2


Interestingly, the frequency of NF2 gene mutation varies between meningioma subtypes. Among the three most common meningioma World Health Organization (WHO) grade I subtypes, 70 to 80% of fibroblastic and transitional meningiomas show NF2 gene mutations, whereas the meningothelial subtype shows NF2 mutations only 25% of the time, suggesting that cytogenetic differences in the tumorigenesis of meningioma subtypes may exist.6 In both atypical and anaplastic meningiomas, the frequency of NF2 gene mutations is ~70%.3 Therefore, NF2 gene mutations are probably involved with tumori-genesis but not tumor progression. In vivo mice experiments have provided further support for this theory but suggest that merlin loss alone is not sufficient for meningioma development.7


A search for a second tumor suppressor gene on 22q stems from the discrepancy between the frequency of chromosome 22 LOH, which exceeds that of NF2 gene abnormalities.6 Deletions of chromosome 22 are found in all NF2-associated meningiomas and in 54 to 78% of sporadic meningiomas. Further analysis of the NF2 gene in sporadic meningiomas reveals that roughly one third to one half of these tumors have an inactivating mutation, often accompanied by loss of the other allele. These studies have resulted in other possible gene candidates ( Fig. 6.3 ; Table 6.1 ).2



Chromosome 1


Deletions of the short arm of chromosome 1 are the second most frequent alteration detected by cytogenetic analysis of meningiomas ( Table 6.1 ).2,3 FISH studies showing monosomy 1p in 70% of atypical and almost 100% of anaplastic meningiomas indicate a correlation between loss of chromosome 1p and meningioma progression. Loss of 1p also correlates with tumor recurrence; the rate of recurrence is 30% with loss of 1p but only 4.3% when 1p is retained.8 Which gene on the 1p arm results in the clinical effects on tumor is unknown, but research has suggested alkaline phosphatase as a possible tumor suppressor whose location on chromosome 1p (1p34→1p36.1) and loss of function are correlated with higher-grade meningiomas ( Table 6.1 ).3,8



Other Meningioma Chromosomal Abnormalities


Many cytogenetic abnormalities are associated with meningioma progression and typical or anaplastic histology. These chromosomal aberrations include the presence of dicentric or ring chromosomes, losses of chromosome arms 1p, 6q, 7, 9p, 10, 14q, 18q, 19, or 20, and gains/amplifications of 1q, 9q, 12q, 15q, 17q, or 20q ( Fig. 6.2 ).3,8,9 It is unknown how these chromosome changes lead to tumor progression, although several chromosomes and genes appear to have specific associations with benign, atypical, and anaplastic meningioma grades. For example, benign meningiomas are more liable to have 14q deletions.3 Roughly two thirds of anaplastic meningiomas exhibit altered cell-cycle checkpoint tumor suppressor genes located on chromosome 9p ( Table 6.1 ). Further evidence implicating these deletions in meningioma pathogenesis comes from the significantly shorter survival times of patients. Other rare molecular abnormalities include phosphatase and tensin homologue gene deletion, cyclin-dependent kinase inhibitor 2c gene deletion, and ribosomal protein S6 kinase gene amplification.1,2


Cytogenetic alterations can also include changes in chromosome number. Sixty percent of meningiomas have been found to be hypodiploid, 33% diploid, 4.5% hyperdiploid, and 2.5% hypotriploid.10 Complex karyo-types with hypodiploidy, structural rearrangements such as ring chromosomes, dicentrics, double minutes, and association between satellites seem to be associated with aggressive tumor characteristics.3 Identification of a microsatellite instability phenotype in meningiomas has also been described. Finally, studies have identified what appear to be sporadic, familial, radiation-induced, and pediatric meningiomas.2,11 Mounting evidence suggests that these tumors are genetically different, which may explain the more aggressive nature of the familial, radiation-induced, and pediatric cases.2,11

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 14, 2020 | Posted by in NEUROLOGY | Comments Off on 6 Molecular Biology of Meningiomas: Tumorigenesis and Growth

Full access? Get Clinical Tree

Get Clinical Tree app for offline access