Skip to main content
  • Research article
  • Open access
  • Published:

Loss of heterozygosity of TRIM3 in malignant gliomas

Abstract

Background

Malignant gliomas are frequent primary brain tumors associated with poor prognosis and very limited response to conventional chemo- and radio-therapies. Besides sharing common growth features with other types of solid tumors, gliomas are highly invasive into adjacent brain tissue, which renders them particularly aggressive and their surgical resection inefficient. Therefore, insights into glioma formation are of fundamental interest in order to provide novel molecular targets for diagnostic purposes and potential anti-cancer drugs. Human Tripartite motif protein 3 (TRIM3) encodes a structural homolog of Drosophila brain tumor (brat) implicated in progenitor cell proliferation control and cancer stem cell suppression. TRIM3 is located within the loss of allelic heterozygosity (LOH) hotspot of chromosome segment 11p15.5, indicating a potential role in tumor suppression. ...

Methods

Here we analyze 70 primary human gliomas of all types and grades and report somatic deletion mapping as well as single nucleotide polymorphism analysis together with quantitative real-time PCR of chromosome segment 11p15.5.

Results

Our analysis identifies LOH in 17 cases (24%) of primary human glioma which defines a common 130 kb-wide interval within the TRIM3 locus as a minimal area of loss. We further detect altered genomic dosage of TRIM3 in two glioma cases with LOH at 11p15.5, indicating homozygous deletions of TRIM3.

Conclusion

Loss of heterozygosity of chromosome segment 11p15.5 in malignant gliomas suggests TRIM3 as a candidate brain tumor suppressor gene.

Peer Review reports

Background

Malignant gliomas are brain tumors that represent devastating and difficult-to-treat cancers with a mean patient survival of 10 months. Although surgical interventions allow resection and improve local tumor control, the further course of the disease remains dominated by re-appearance of unscheduled cell proliferation and insidious infiltration of normal brain tissue. Especially glioblastomas appear notoriously resistant to therapy, which has been attributed to DNA-repair proficiency and deregulated molecular pathways [1, 2]. More recently combined chemoradiotherapy of concomitant and adjuvant temozolomide and radiotherapy has been introduced which can lead to significant prolongation of survival, in particular in patients with an epigenetically silenced DNA repair gene [3]. However, the outcome remains poor. This therapeutic resistance has recently been attributed to tumor stem-like cells due to their unrestrained self-renewal capacity and the ability to maintain tumorigenic potential at the single cell level, thereby evading both resection and radiotherapy [4–7]. There is growing evidence that some brain cancers arise either from normal stem cells or from progenitor cells in which self-renewal pathways have become aberrantly activated [7–10].

Drosophila Brain tumor (brat) has been identified as a regulator of progenitor cell proliferation control and cancer stem cell suppression [11–18]. Brat is expressed throughout Drosophila brain development and exerts an essential gate-keeper function in the binary switch between self-renewal and differentiation of neural progenitor cells. Neural progenitor cells mutated for Brat are unable to differentiate but rather continue to proliferate, resulting in brat mutant cells that display characteristic features of cancer-like stem cells. The resulting brain tumor tissue is characterized by pleiomorphic cells, continued proliferation and chromosome instability, as evidenced by a variety of karyotypic abnormalities [19].

Homologues of brat have been recorded in various species, with three human homologues, namely Tripartite Motif Protein 2 (TRIM2), TRIM3, and TRIM32, located on chromosome 4q31.3, 11p15.5, and 9q33.1, respectively [20–23]. 11p15 represents the telomeric end of chromosome 11 which shows loss of allelic heterozygosity (LOH) in various types of tumors, indicating the presence of one or more tumor suppressor genes [24–27]. Homologues of TRIM3 are primarily expressed in brain and may function at the interface of proliferation and differentiation during the maturation of brain tissue [28–30]. Here we report refined deletion mapping of chromosome 11p15.5 in malignant gliomas.

Methods

Biopsies and DNA extraction

Tumor samples used in this study were obtained from 70 patients who underwent glioma resection at the University Hospital of Basel between 1996 and 2005. The collection of tumor samples has been approved by the Ethics Committee of Basel-Land and Basel-Stadt (EKBB). Informed consent has been obtained together with the patient's permission to conduct open brain surgery, consenting to the use of biopsies for anonymous scientific research. This procedure follows the present recommendations of the Swiss Academy of Medical Sciences as proposed in 2008 and is in compliance with the Helsinki Declaration. Tumors were classified according to the revised WHO classification of tumors of the nervous system [31], comprising 10 oligodendrogliomas grade II, 13 astrocytomas grades I to III, and 47 glioblastoma multiforme [see Additional file 1]. DNA was extracted from fresh frozen primary gliomas and peripheral blood mononuclear cells (PBMCs) derived from the same patients, as previously described [32, 33]. Only material containing less than 30% residual amounts of non-neoplastic cells was considered for further analysis.

STS- and SNP-based LOH analysis

Sequence tagged site (STS)-based LOH was performed essentially as described [32]. Briefly, DNA from 70 glioma specimens and PBMCs of the same patients was analysed for loss of heterozygosity by amplification of microsatellite sequences [32]. Primers for these sequences were obtained from Microsynth (Balgach, Switzerland). Fluorescence based LOH mapping was employed with DNA from all gliomas. D11S4905 (located in TRIM3 intron 2) and D11S1250 primers were FAM-labeled, D1S1318 and D11S1758 primers were HEX-labeled, and D11S1331 and D11S1997 primers were TET-labeled. PCR product size fractionation and quantification were performed on ABI Prism 310 Genetic Analyzer (PE Applied Biosystems, Foster City, CA, USA). The ratio of peak heights of both alleles was calculated for each tumor and PBMC DNA sample. For informative cases, allelic loss was scored if the ratio between tumor and PBMC DNA was more than 1.5 (1/0) or less than 0.66 (0/2). Single nucleotide polymorphism (SNP)-based LOH was performed with the following markers: rs11605881 (TRIM3 promoter), rs11607224 (exon1), rs1060067 (exon4) rs16913748 (exon6), rs11605141 (intron6), rs13343175 (intron7), rs3830325 (intron9), rs2306897 (exon10) and rs2723636 (exon13). SNPs were visualized on an ABI Prism 310 Genetic Analyzer (PE Applied Biosystems, Foster City, CA, USA).

Quantitative real-time PCR

Classification of the genetic status of TRIM3 was performed by quantitative real-time-PCR using the TaqMan ABI Prism® 7700 Sequence Detection System (PE Applied Biosystems, Foster City, CA, USA) as previously described [34]. Gene-specific primers for the reference-control GAPDH, which is located on chromosome 12p13.3, as well as primers for Intron1, Exon 3, Intron 6, Intron 11 and cDNA of TRIM3 were used as follows:

For GAPDH:

5'-AATGGGACTGAGGCTCCAC (sense),

5'-TTATGGGAAAGCCAGTCCCC (antisense).

For TRIM3 Intron 1:

5'-CCCCAAGGGTGCGTTTGTATT (sense),

5'-TGCTCTCACGGACATGGACA (antisense).

For TRIM3 Exon 3:

5'-GCAGTTCCTGGTATGCAGCAT (sense),

5'-TGCAGGCAAGGAAGAACCTT (antisense).

For TRIM3 Intron 6:

5'-GGGCCAAACAGAAGGTGTGT (sense),

5'-GGCATGTCAGGAGGCAGAAT (antisense).

For TRIM3 Intron 11:

5'-AGGCAGTAGGGCACATGGAT (sense),

5'-GAGAACCCCCACCCAGATCT (antisense).

For TRIM3 cDNA Exons 7/8:

5'-GGCGGCAAACGAAAGGA (sense),

5'-CCTTCCACGACTGCCAACA (antisense).

Gene-specific double-dye FAM-TAMRA labelled oligomeric probes were:

For GAPDH: FAM-ATCCAAGACTGGCTCCTCCCTGCTG-TAMRA.

For TRIM3 Intron 1: FAM-CCCACAGCCGCTCCGACCCA-TAMRA.

For TRIM3 Exon 3: FAM-TGCCTGGATCGGTACCAGTGCCC-TAMRA.

For TRIM3 Intron 6: FAM-CACCAGCTCCCCATTCCCCA-TAMRA.

For TRIM3 Intron 11: FAM-CAGCTACAGCCCAAATCTGCTTCATAGGCTT-TAMRA.

For TRIM3 cDNA: FAM-AACCCAATTGAGGATGAGCTCGTCTTCC-TAMRA.

PCR conditions, primers and probe design were assessed by the Primer Express® program (PE Applied Biosystems, Foster City, CA, USA). Final primer concentration was 200 nM and fluorescent oligomeric probe concentration was 50 nM. Fifty ng genomic DNA derived from primary tumor tissue was taken as template. Genomic DNA amounts between normal and neoplastic tissues were standardized by subtracting the respective threshold cycles (Cts) obtained for the GADPH gene. In this approach the variable Ct is defined as the fractional cycle number crossing a fixed threshold of fluorescence that has been generated by cleavage of the probe due to polymerase driven exonucleolytic activity. Differences in the Ct values between two genes were referred to the ΔCt value. After normalization of the tumor Ct values with the PBMC Ct values, the ΔCtvalues and the relative copy number of Introns1, 6 and 11, as well as Exon 3 of TRIM3 were calculated as follows: ΔCt (X) = Ct (reference) - Ct (X). Thus, ΔCt values of 0 ± 0.3 [-0.3;+0.3] indicated retention of both alleles (diploidy = 2n) and ΔCt values of -1 ± 0.3 [-0.7;-1.3] indicated loss of one allele (haploidy = n). Due to the frequent contamination of tumor biopsy DNA by that of invaded normal tissue, ΔCt values of < -1.30 were accounted as indicative for homozygous deletions. All analyses of tumor samples were performed in triplicate in parallel with PBMC DNA, and data indicating homozygous deletions resulted from two independent experiments.

Results

LOH in human gliomas reveals a 130 kb minimally lost area uncovering TRIM3

Initial studies had identified an approximately 21 centiMorgan region on 11p15.5-pter that showed frequent loss of heterozygosity in malignant gliomas [35]. This region has been refined to 7 megabases (Mb) [24], spanning the region 11p15.4–5 between microsatellite markers (also called STS) D11S922 and D11S1250 (Fig. 1). Several genes are contained within this interval that might be involved in brain development and/or tumor suppression, including TRIM3 and a cluster of genes encoding the more distantly related TRIM5, TRIM6, TRIM22, and TRIM34 genes. This area also contains ASCL2 and ASCL3. Both genes are homologues of the Drosophila genes of the achaete-scute (ASC) complex that promote cells to develop a neural fate [36], which therefore may represent additional candidate brain tumor suppressors encoded by this region (Fig. 1). We performed loss of heterozygosity analysis on human brain tumor samples representing 70 primary gliomas of varying histology and grades including 13 astrocytomas (AS) WHO grades I to III, 10 oligodendrogliomas (OG) WHO grade II, and 47 glioblastomas (GBM). We focused our LOH analysis on the region spanning TRIM3 by using 6 microsatellite markers in addition to the ones that had been used earlier to determine the minimal area of loss [24].

Figure 1
figure 1

Deletion mapping of chromosomal region 11p15.5 in human primary gliomas defines a 130 kb-wide area containing TRIM3. A. The 11p15.5 chromosomal region highlighting TRIM3, the more distantly related TRIM5, TRIM6, TRIM22, and TRIM34 genes, as well as ASCL2 and ASCL3; arrows indicate transcription orientations. Middle panel: somatic deletion of the 11p15.5 region in glioma. Tumor histology, grades and numbers are indicated on the left. Allelic retention, allelic loss and non-informative data are represented by open, closed and grey circles, respectively. Minimal and maximal extensions of allelic loss are shown with filled and hatched grey areas, respectively. The 11p15 microsatellite markers/sequence tag sites (STS) used are indicated (D11S1318, D11S1758, D11S1997, D11S4905, D11S1331, D11S1250). Alignment of LOH data delimits a minimal area of 130 kb showing LOH common to all 10 tumors between markers D11S1997 and D11S4905 which identifies the TRIM3 locus. GBMs displaying complete loss of the central region were not tested for D11S1318 and D11S1250 (ND). B. Informative LOH data with marker D11S4905 comparing normal (N) with tumor (T) DNA for brain tumors OGII 040, ASIII 098, ASII 141, GBM 157, GBM 164. Abbreviations: GBM, Glioblastoma multiforme; AS, astrocytoma; OG, oligodendroglioma; RET, retention; LOH, loss of heterozygosity; ND, not determined; NI, non-informative.

Among the 70 primary tumor samples tested, 17 out of 70 (24%) showed LOH of at least one marker in this region. Heterozygous deletions included 12 GBM out of 47 analysed (25%) consistent with the 11p15.5 deletion frequency previously described in GBM [24]. Further, one OG grade II out of 10 (10%) showed 11p15.5 allelic loss. No allelic loss was detected in AS grade I (0/3), while heterozygous deletions were observed in 2 AS grade II out of 7 (28%) as well as in 2 AS grade III out of 3 (67%), suggesting a graded escalation of 11p15.5 deletion frequency with tumor grading in AS (see Table 1 and Fig. 1). Some of these heterozygous deletions, including OGII 040, ASII 141, and GBM 157, covered the complete region 11p15 (D11S1318, D11S1758, D11S1997, D11S4905, D11S1331, D11S1250). At the telomeric side, GBM tumor 167 showed the longest retention proximally extending to STS marker D11S1997, whereas ASII tumor 031 had the most distal extension of retention to STS marker D11S4905. Therefore, alignment of our LOH data with the physical map of the 11p telomeric region delimited a minimal area of loss common to all of these tumors between markers D11S1997 and D11S4905. This interval reduced the minimal area of loss from 7 Mb to only 130 kilobases (kb) covering the TRIM3 locus (Fig. 1), and also pointed to potential breakpoint mutations within the TRIM3 gene between Exons 3–13.

Table 1 11p15.5 LOH frequencies among glioma subsets

Genomic dosage alterations of TRIM3in malignant gliomas

In order to refine somatic deletion mapping and to delimit the minimal area of loss in more detail, we used single nucleotide polymorphic (SNP) markers located within the TRIM3 genomic area to further investigate a selection of tumor samples. For further analysis, we selected those tumors that were indicative for loss or at least partial loss of the analysed region at 11p15, namely GBMs 149, 157, 164, 167, and 211, as well as ASII 031, ASIII 023, and ASIII 098 (Fig. 2A).

Figure 2
figure 2

SNP-based somatic deletion mapping of chromosomal region 11p15.5 identifies potential breakpoint mutations within the TRIM3 gene. A. Superimposition of single nucleotide polymorphism (SNP) and sequence tag site (STS)-based LOH data. (Note that compared to Fig. 1, the map has been inverted to comply with the 5'-3' transcription orientation of the TRIM3 gene). STS markers (D11S series) are indicated on the top; tumor histology, grades and numbers are indicated on the left. Allelic retention, allelic loss and non-informative data are represented by open, closed and grey circles, respectively. Minimal and maximal extension of STS-based areas of allelic loss described in Fig. 1, are shown with filled and hatched grey areas, respectively. Areas of local allelic retention defined by SNP analysis within segments of allelic loss defined by STS analysis are framed by open rectangles. SNP and STS data, as well as SNP markers (rs series) used are related to a structural map of the TRIM3 gene with its coding regions shown in black (bottom). Thus, SNP-based allelic retention of short sections within the areas of LOH in primary gliomas ASIII 098, GBM 157, and GBM 164 suggest potential homozygous deletions within the TRIM3 gene. B. SNP bi-allelism in primary tumors with 11p15.5 allelic loss. Sequence electrophoretograms of genomic DNA extracted from primary tumor samples amplified at indicated SNP markers of the TRIM3 gene area; peak color codes: green (A), blue (C), black (G), red (T). Abbreviations: GBM, Glioblastoma multiforme; AS, astrocytoma; OG, oligodendroglioma; RET, retention; LOH, loss of heterozygosity; NI, non-informative.

Among the ten SNPs initially selected, six turned out to be non-informative in all analyzed tumors whereas four SNPs, namely rs11605881, rs11607224, rs16913748, rs11605141, displayed loss of heterozygosity or allelic retention in those tumors selected for further analysis (Fig. 2). Thus, we observed allelic retention of both parental alleles of SNPs rs11605881, rs16913748, and rs11605141 in primary tumors ASII 031 and GBM 211, respectively. These data displaced the centromeric rim of the minimally lost area of TRIM3 from STS D11S4905 to SNP rs11605141 but still targeted the TRIM3 gene (Fig. 2). In addition, in those cases where we observed STS-based loss of heterozygosity extending on both sides of the TRIM3 gene (ASIII 098, GBM 157 and GBM 164), the detected area of LOH was locally interrupted by short sections with allelic retention at SNPs rs11605881 and rs11607224. Indeed, tumors ASIII 098 and GBM 164 showed heterozygosity at SNP markers rs11605881 and rs11607224 located in the TRIM3 promoter and in exon1, respectively, whereas analysis of markers rs16913748 and rs11605141 of TRIM3 intron 6 revealed heterozygosity in GBM 157 (Fig. 2).

Allelic retention within a chromosomal interval displaying LOH has been interpreted as a potential site of homozygous deletion, where retention seems to result from the amplification of wildtype DNA deriving from non-neoplastic cells present in the tumor biopsy [37]. Thus, SNP-based allelic retention of short sections within the areas of LOH in primary gliomas ASIII 098, GBM 157, and GBM 164 indicated potential homozygous deletions within the TRIM3 gene. In order to investigate this possibility, we targeted four equidistant regions of the TRIM3 gene, including the two areas of possible homozygous loss in the three primary tumor samples ASIII 098, GBM 157 and GBM 164 by quantitative real-time PCR (Q-PCR). Analysis of the genetic status of TRIM3 in ASIII 098, GBM 157, and GBM 164 was assayed on DNA extracted from both the primary gliomas and peripheral blood mononuclear cells (PBMCs) derived from the same patients by Q-PCR of the reference-control gene GAPDH, as well as for Intron1, Exon 3, Intron 6, and Intron 11 of TRIM3, respectively (see Methods).

In ASIII 098, tumor genomic dosage in TRIM3 intron 1, at the site between SNP markers rs11605881 and rs11607224 indicated DNA levels below haploidy of ΔCt = -1.98 ± 0.40 (for calculation details, see Methods). These results signified a homozygous deletion encompassing TRIM3 intron 1 as already indicated by data of SNP rs11605881 analysis (Fig. 3). Similarly, Q-PCR analysis of GBM 157 indicated DNA levels below haploidy from TRIM3 intron 6 (ΔCt = -1.59 ± 0.20) to intron 11 (ΔCt = -1.43 ± 0.20), signifying homozygous deletion as already indicated by SNPs markers rs1693748 and rs1605141 (Fig. 3). In contrast, genomic dosage of primary tumor GBM 164 indicated continuous diploidy along the TRIM3 gene as exemplified by ΔCt values between -0.30 and +0.30 of four Q-PCR markers covering intron 1, exon 3, intron 6 and intron 11 (Fig. 3). Thus, among the 10 primary human glioma identified with allelic loss at 11p15.5, Q-PCR analysis of ASIII 098 and GBM 157 (20%) indicated homozygous deletions within the TRIM3 gene.

Figure 3
figure 3

Altered genomic dosage of TRIM3 suggests homozygous deletions in human primary gliomas. A. Genomic dosage of the TRIM3 gene area of ASIII 098, GBM 157, and GBM 164 as determined by quantitative real-time PCR. Light grey background indicates ΔCt cut-offs for diploidy (2n = 0 ± 0.3) and haploidy (n = -1 ± 0.3) for each primary tumor sample; grey bars depict ΔCt values of amplified tumor DNA related to a structural map of the TRIM3 gene with coding regions shown in black B. Mapping of indicated homozygous deletions within the TRIM3 gene. STS-based and SNP-based compilation of Q-PCR data is shown in relation to structural map of the TRIM3 gene with coding regions shown in black. Tumor histology, grades and numbers are indicated on the left. STS-based (D11S series) allelic loss and non-informative data are represented by closed and grey circles, respectively. Areas of local allelic retention defined by SNP analysis (rs series) within segments of allelic loss defined by STS analysis are framed by open rectangles. Copy number deduced from Q-PCR data are indicated; minimal and maximal extension of allelic loss' are shown with filled and hatched grey areas, respectively; areas of homozygous loss are shown in black. Q-PCR data indicate homozygous deletions in primary tumors ASIII 098 and GBM 157. Abbreviations: GBM, Glioblastoma multiforme; AS, astrocytoma; OG, oligodendroglioma; RET, retention; LOH, loss of heterozygosity; NI, non-informative; Q-PCR, quantitative real-time polymerase chain reaction.

Discussion

In cancer research, conventional strategies of somatic deletion mapping rely on the detection of frequent sites of larger DNA alterations. Accordingly, allelic loss of heterozygosity analysis of tumor DNA facilitates the identification and localization of a minimally lost area correlating with candidate tumor suppressor gene loci that might be involved in the tumorigenic process [38]. In contrast to these conventional top-down strategies, we carried out somatic deletion mapping on human glioma DNA focusing on the TRIM3 locus. We opted for this unorthodox bottom-up strategy as a result of significant structural homologies between TRIM3 and the Drosophila brain tumor suppressor Brat [19], as well as owing to the fact that TRIM3 is located on chromosome segment 11p15.5.

Previous analyses demonstrated that chromosome segment 11p15 contains a region of frequent loss of allelic heterozygosity in various adult tumors including those of the brain, lung, breast, ovary, esophagus, stomach, as well as others [27, 35, 39–44]. The frequency of LOH in this region and its apparent correlation with metastatic tumor spread suggests that this chromosome segment may represent a hotspot containing one or more tumor suppressor gene(s).

In the case of brain tumors, a previous LOH study delimited the minimal area of loss to a final 7 Mb-wide genomic interval spanning several genes with a potential role in both developing and neoplastic brain tissue [24]. Among those genes are ASCL2 and ASCL3, mammalian homologues of Drosophila achaete-scute complex [36], as well as TRIM3, a homologue of Drosophila brain tumor involved in progenitor cell proliferation control and cancer stem cell suppression [19], and a cluster of more distantly related TRIM genes (see Figure 1).

Compared to the earlier study by Schiebe et al. [24], we analyzed an equivalent number of GBMs (n = 47 vs. n = 50), as well as brain tumors that are distinct in origin, i.e. OG (n = 10) and AS (n = 13). By focusing on GBM only (see Table 1), our data revealed similar deletion frequencies (25% vs. 28%) to those previously obtained in GBM [24]. Interestingly, our deletion analysis revealed increased frequencies in AS of higher grade, which might indicate a possible association of 11p15.5 allelic loss with tumor progression. However, this conclusion needs to be strengthened by the analysis of a larger number of AS.

Furthermore, as compared to the 7 Mb region previously described [24], our somatic deletion mapping analysis delimited a 130 kb-wide minimal area of loss. Thus, our results rule out ASCL2 and ASCL3 and the more distantly related TRIM genes as potential glioma tumor suppressors within the genomic region analyzed. Significantly, the 130 kb-wide minimal area of loss not only identified the TRIM3 locus, but also indicated potential breakpoint mutations within the TRIM3 gene.

We further substantiated our LOH data by single nucleotide polymorphism analysis together with quantitative real-time PCR. Previous studies showed that Q-PCR can identify micro-deletions providing a reliable approach for a direct and specific determination of the ploidy status within defined genetic loci. This approach led to the identification of homozygous deletions of the p14ARF/p16INK4a tumor suppressor locus which is frequently affected in human gliomas [34]. Significantly, we detected genomic dosage alterations of TRIM3 in two glioma cases with LOH at 11p15.5, indicating homozygous deletions of TRIM3. Our LOH and Q-PCR data therefore suggest that TRIM3 may act as a tumor suppressor in the human brain. However, in vitro and mammalian in vivo loss- as well- as gain-of function analyses are required to determine the function of TRIM3 in detail. It will be interesting to see whether TRIM3, similar to its Drosophila homologue Brain tumor, is involved in the regulation of progenitor cell proliferation control and brain tumor suppression.

Conclusion

Our analysis identifies loss of allelic heterozygosity at 11p15.5 in 17 cases of primary human glioma and defines a common 130 kb-wide interval as a minimal area of loss that covers the TRIM3 locus. In two glioma cases with LOH, altered genomic dosage of TRIM3 indicates homozygous deletions. Together, these data suggest TRIM3 as a 11p15.5 candidate brain tumor suppressor gene. Further investigation will be needed to elucidate the biological function of TRIM3 and its precise role in brain tumor suppression.

Abbreviations

GBM:

Glioblastoma multiforme

AS:

astrocytoma

OG:

oligodendroglioma

RET:

retention

LOH:

loss of heterozygosity

ND:

not determined

NI:

non-informative

Q-PCR:

quantitative real-time polymerase chain reaction.

References

  1. Merlo A: Genes and pathways driving glioblastomas in humans and murine disease models. Neurosurg Rev. 2003, 26: 145-158.

    Article  PubMed  Google Scholar 

  2. The Cancer Genome Atlas Research Network: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008, 455: 1061-1068.

    Article  Google Scholar 

  3. Stupp R, Hegi ME, Gilbert MR, Chakravarti A: Chemoradiotherapy in malignant glioma: Standard of care and future directions. J Clin Oncol. 2007, 25: 4127-4136.

    Article  CAS  PubMed  Google Scholar 

  4. Al-Hajj M, Clarke MF: Self-renewal and solid tumor stem cells. Oncogene. 2004, 23: 7274-7282.

    Article  CAS  PubMed  Google Scholar 

  5. Sanai N, Alvarez-Buylla A, Berger MS: Neural stem cells and origin of gliomas. N Engl J Med. 2005, 353: 811-822.

    Article  CAS  PubMed  Google Scholar 

  6. Vescovi AL, Galli R, Reynolds BA: Brain tumor stem cells. Nat Rev Cancer. 2006, 6: 425-236.

    Article  CAS  PubMed  Google Scholar 

  7. Stiles CD, Rowitch DH: Glioma stem cells: A midterm exam. Neuron. 2008, 58: 832-846.

    Article  CAS  PubMed  Google Scholar 

  8. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB: Identification of human brain tumour initiating cells. Nature. 2004, 432: 396-401.

    Article  CAS  PubMed  Google Scholar 

  9. Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P, Magdaleno S, Dalton J, Calabrese C, Board J, Macdonald T, Rutka J, Guha A, Gajjar A, Curran T, Gilbertson RJ: Radial glia cells are candidate stem cells of ependymoma. Cancer Cell. 2005, 8: 323-335.

    Article  CAS  PubMed  Google Scholar 

  10. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN: Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006, 444: 756-760.

    Article  CAS  PubMed  Google Scholar 

  11. Arama E, Dickman D, Kinchie Z, Shearn A, Lev Z: Mutations in the beta-propeller domain of the Drosophila brain tumor (brat) protein induce neoplasm in the larval brain. Oncogene. 2000, 19: 3706-3716.

    Article  CAS  PubMed  Google Scholar 

  12. Loop T, Leemans R, Stiefel U, Hermida L, Egger B, Xie F, Primig M, Certa U, Fischbach KF, Reichert H, Hirth F: Transcriptional signature of an adult brain tumor in Drosophila. BMC Genomics. 2004, 5: 24-

    Article  PubMed  PubMed Central  Google Scholar 

  13. Caussinus E, Gonzalez C: Induction of tumor growth by altered stem-cell asymmetric division in Drosophila melanogaster. Nat Genet. 2005, 37: 1125-1129.

    Article  CAS  PubMed  Google Scholar 

  14. Bello B, Reichert H, Hirth F: The brain tumor gene negatively regulates neural progenitor cell proliferation in the larval central brain of Drosophila. Development. 2006, 133: 2639-2648.

    Article  CAS  PubMed  Google Scholar 

  15. Lee CY, Wilkinson BD, Siegrist SE, Wharton RP, Doe CQ: Brat is a Miranda cargo protein that promotes neuronal differentiation and inhibits neuroblast self-renewal. Dev Cell. 2006, 10: 441-449.

    Article  CAS  PubMed  Google Scholar 

  16. Betschinger J, Mechtler K, Knoblich JA: Asymmetric segregation of the tumor suppressor Brat regulates self-renewal in Drosophila neural stem cells. Cell. 2006, 124: 1241-1253.

    Article  CAS  PubMed  Google Scholar 

  17. Beaucher M, Goodliffe J, Hersperger E, Trunova S, Frydman H, Shearn A: Drosophila brain tumor metastases express both neuronal and glial cell type markers. Dev Biol. 2007, 301: 287-297.

    Article  CAS  PubMed  Google Scholar 

  18. Bowman SK, Rolland V, Betschinger J, Kinsey KA, Emery G, Knoblich JA: The tumor suppressors Brat and Numb regulate transit-amplifying neuroblast lineages in Drosophila. Dev Cell. 2008, 14: 1-12.

    Article  Google Scholar 

  19. Caussinus E, Hirth F: Asymmetric cell division in development and cancer. Prog Mol Subcell Biol. 2007, 45: 205-225.

    Article  CAS  PubMed  Google Scholar 

  20. Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, Riganelli D, Zanaria E, Messali S, Cainarca S, Guffanti A, Minucci S, Pelicci PG, Ballabio A: The tripartite motif family identifies cell compartments. EMBO J. 2001, 20: 2140-2151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. El-Husseini AE, Fretier P, Vincent SR: Cloning and characterization of a gene (RNF22) encoding a novel brain expressed ring finger protein (BERP) that maps to human chromosome 11p15.5. Genomics. 2001, 71: 363-367.

    Article  CAS  PubMed  Google Scholar 

  22. Frosk P, Weiler T, Nylen E, Sudha T, Greenberg CR, Morgan K, Fujiwara TM, Wrogemann K: Limb-girdle muscular dystrophy type 2H associated with mutation in TRIM32, a putative E3-ubiquitin-ligase gene. Am J Hum Genet. 2002, 70: 663-672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sardiello M, Cairo S, Fontanella B, Ballabio A, Meroni G: Genomic analysis of the TRIM family reveals two groups of genes with distinct evolutionary properties. BMC Evol Biol. 2008, 8: 225-

    Article  PubMed  PubMed Central  Google Scholar 

  24. Schiebe M, Ohneseit P, Hoffmann W, Meyermann R, Rodemann HP, Bamberg M: Loss of heterozygosity at 11p15 and p53 alterations in malignant gliomas. J Cancer Res Clin Oncol. 2001, 127: 325-328.

    Article  CAS  PubMed  Google Scholar 

  25. Scelfo RA, Schwienbacher C, Veronese A, Gramantieri L, Bolondi L, Querzoli P, Nenci I, Calin GA, Angioni A, Barbanti-Brodano G, Negrini M: Loss of methylation at chromosome 11p15.5 is common in human adult tumors. Oncogene. 2002, 21: 2564-2572.

    Article  CAS  PubMed  Google Scholar 

  26. Lam CT, Tang CM, Lau KW, Lung ML: Loss of heterozygosity on chromosome 11 in esophageal squamous cell carcinomas. Cancer Lett. 2002, 178: 75-81.

    Article  CAS  PubMed  Google Scholar 

  27. Larson PS, Schlechter BL, King CL, Yang Q, Glass CN, Mack C, Pistey R, de Las Morenas A, Rosenberg CL: CDKN1C/p57kip2 is a candidate tumor suppressor gene in human breast cancer. BMC Cancer. 2008, 8: 68-

    Article  PubMed  PubMed Central  Google Scholar 

  28. El-Husseini AE, Vincent SR: Cloning and characterization of a novel RING finger protein that interacts with class V myosins. J Biol Chem. 1999, 274: 19771-1977.

    Article  CAS  PubMed  Google Scholar 

  29. El-Husseini AE, Kwasnicka D, Yamada T, Hirohashi S, Vincent SR: BERP, a novel ring finger protein, binds to alpha-actinin-4. Biochem Biophys Res Commun. 2000, 267: 906-911.

    Article  CAS  PubMed  Google Scholar 

  30. Van Diepen MT, Spencer GE, van Minnen J, Gouwenberg Y, Bouwman J, Smit AB, van Kesteren RE: The molluscan RING-finger protein L-TRIM is essential for neuronal outgrowth. Mol Cell Neurosci. 2005, 29: 74-81.

    Article  CAS  PubMed  Google Scholar 

  31. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC, Cavenee WK: The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol. 2002, 61: 215-225.

    Article  PubMed  Google Scholar 

  32. Maier D, Comparone D, Taylor E, Zhang Z, Gratzl O, Van Meir EG, Scott RJ, Merlo A: New deletion in low-grade oligodendroglioma at the glioblastoma suppressor locus on chromosome 10q25–26. Oncogene. 1997, 15: 997-1000.

    Article  CAS  PubMed  Google Scholar 

  33. Maier D, Zhang Z, Taylor E, Hamou MF, Gratzl O, Van Meir EG, Scott RJ, Merlo A: Somatic deletion mapping on chromosome 10 and sequence analysis of PTEN/MMAC1 point to the 10q25–26 region as the primary target in low-grade and high-grade gliomas. Oncogene. 1998, 16: 3331-3335.

    Article  CAS  PubMed  Google Scholar 

  34. Labuhn M, Jones G, Speel EJ, Maier D, Zweifel C, Gratzl O, Van Meir EG, Hegi ME, Merlo A: Quantitative real-time PCR does not show selective targeting of p14(ARF) but concomitant inactivation of both p16(INK4A) and p14(ARF) in 105 human primary gliomas. Oncogene. 2001, 20: 1103-1109.

    Article  CAS  PubMed  Google Scholar 

  35. Sonoda Y, Iizuka M, Yasuda J, Makino R, Ono T, Kayama T, Yoshimoto T, Sekiya T: Loss of heterozygosity at 11p15 in malignant glioma. Cancer Res. 1995, 55: 2166-2168.

    CAS  PubMed  Google Scholar 

  36. Campuzano S, Modolell J: Patterning of the Drosophila nervous system: the achaete-scute gene complex. Trends Genet. 1992, 8: 202-208.

    Article  CAS  PubMed  Google Scholar 

  37. Cairns P, Tokino K, Eby Y, Sidransky D: Homozygous deletions of 9p21 in primary human bladder tumors detected by comparative multiplex polymerase chain reaction. Cancer Res. 1994, 54: 1422-1424.

    CAS  PubMed  Google Scholar 

  38. Ohgaki H, Kleihues P: Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 2007, 170: 1445-1453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Baffa R, Negrini M, Mandes B, Rugge M, Ranzani GN, Hirohashi S, Croce CM: Loss of heterozygosity for chromosome 11 in adenocarcinoma of the stomach. Cancer Res. 1996, 56: 268-272.

    CAS  PubMed  Google Scholar 

  40. Fults D, Petronio J, Noblett BD, Pedone CA: Chromosome 11p15 deletions in human malignant astrocytomas and primitive neuroectodermal tumors. Genomics. 1992, 14: 799-801.

    Article  CAS  PubMed  Google Scholar 

  41. Hoffmann MJ, Florl AR, Seifert HH, Schulz WA: Multiple mechanisms downregulate CDKN1C in human bladder cancer. Int J Cancer. 2005, 114: 406-413.

    Article  CAS  PubMed  Google Scholar 

  42. Lu KH, Weitzel JN, Kodali S, Welch WR, Berkowitz RS, Mok SC: A novel 4-cM minimally deleted region on chromosome 11p15.1 associated with high grade nonmucinous epithelial ovarian carcinomas. Cancer Res. 1997, 57: 387-90.

    CAS  PubMed  Google Scholar 

  43. Nakata T, Yoshimoto M, Kasumi F, Akiyama F, Sakamoto G, Nakamura Y, Emi M: Identification of a new commonly deleted region within a 2-cM interval of chromosome 11p11 in breast cancers. Eur J Cancer. 1998, 34: 417-421.

    Article  CAS  PubMed  Google Scholar 

  44. Zhao B, Bepler G: Transcript map and complete genomic sequence for the 310 kb region of minimal allele loss on chromosome segment 11p15.5 in non-small-cell lung cancer. Oncogene. 2001, 20: 8154-8164.

    Article  CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Acknowledgements

We thank Mihai Ionescu and Heinrich Reichert for comments on earlier versions of the manuscript, and Heinrich Reichert for support. This work was funded by grants from the Swiss Cancer League (OCS-01613-12-2004) and the Krebsliga beider Basel (No. 7-2004) (to FH and AM).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Adrian Merlo or Frank Hirth.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

FH conceived, and FH, JLB and AM designed the study; US, ET, and BD acquired the data; FH, JLB and AM analyzed and interpreted the data; FH and JLB wrote the manuscript. All authors read and approved the final manuscript.

Electronic supplementary material

12885_2008_1405_MOESM1_ESM.pdf

Additional file 1: LOH data for all gliomas investigated. The data provided represent all 70 glioma cases investigated.(PDF 27 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Boulay, JL., Stiefel, U., Taylor, E. et al. Loss of heterozygosity of TRIM3 in malignant gliomas. BMC Cancer 9, 71 (2009). https://doi.org/10.1186/1471-2407-9-71

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2407-9-71

Keywords