The emerging role of NG2 in pediatric diffuse intrinsic pontine glioma

Sridevi Yadavilli, Joseph Scafidi, Oren J Becher, Amanda M Saratsis, Rebecca L Hiner, Madhuri Kambhampati, Santi Mariarita, Tobey J MacDonald, Kari-Elise Codispoti, Suresh N Magge, Jyoti K Jaiswal, Roger J Packer, Javad Nazarian, Sridevi Yadavilli, Joseph Scafidi, Oren J Becher, Amanda M Saratsis, Rebecca L Hiner, Madhuri Kambhampati, Santi Mariarita, Tobey J MacDonald, Kari-Elise Codispoti, Suresh N Magge, Jyoti K Jaiswal, Roger J Packer, Javad Nazarian

Abstract

Diffuse intrinsic pontine gliomas (DIPGs) have a dismal prognosis and are poorly understood brain cancers. Receptor tyrosine kinases stabilized by neuron-glial antigen 2 (NG2) protein are known to induce gliomagenesis. Here, we investigated NG2 expression in a cohort of DIPG specimens (n= 50). We demonstrate NG2 expression in the majority of DIPG specimens tested and determine that tumors harboring histone 3.3 mutation express the highest NG2 levels. We further demonstrate that microRNA 129-2 (miR129-2) is downregulated and hypermethylated in human DIPGs, resulting in the increased expression of NG2. Treatment with 5-Azacytidine, a methyltransferase inhibitor, results in NG2 downregulation in DIPG primary tumor cells in vitro. NG2 expression is altered (symmetric segregation) in mitotic human DIPG and mouse tumor cells. These mitotic cells co-express oligodendrocyte (Olig2) and astrocyte (glial fibrillary acidic protein, GFAP) markers, indicating lack of terminal differentiation. NG2 knockdown retards cellular migration in vitro, while NG2 expressing neurospheres are highly tumorigenic in vivo, resulting in rapid growth of pontine tumors. NG2 expression is targetable in vivo using miR129-2 indicating a potential avenue for therapeutic interventions. This data implicates NG2 as a molecule of interest in DIPGs especially those with H3.3 mutation.

Keywords: DIPG; NG2; PDGF; glioma; histone 3.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors declare no potential conflicts of interest.

Figures

Figure 1. Immunohistochemical and western blotting analysis…
Figure 1. Immunohistochemical and western blotting analysis validates NG2 upregulation in human DIPG
(a) In human tissue, diffuse expression of NG2 was observed in DIPG tumor localized to membrane (inset). (b) Adjacent healthy brainstem tissue. Scale bar: 100 μM. (c) Graphical representation of western blotting data of NG2 expression in 22 DIPG tumor and 16 adjacent normal specimens. GAPDH was used for protein loading normalization. Average NG2 expression was plotted after normalizing NG2 levels to GAPDH in each sample. P < 0.05 was considered significant. (d) Average of normalized NG2 expression levels in NG2 western blot samples with histone 3.1 K27M mutation or histone 3.3 K27M mutation. P < 0.05 was considered significant.
Figure 2. NG2 overexpression in PDGFB and…
Figure 2. NG2 overexpression in PDGFB and PDGFB/H3.3 K27M/p53−/− mouse models
(a) In mouse, expression of NG2 in PDGFB mouse was limited to brainstem tumor (dotted area). (b) Sporadic expression of NG2 across brainstem and cerebellum of normal mouse. (c) Injection of NSG SCID mouse with mouse DIPG cells that are H3.3.K27 mutant and have PDGFB overexpression and p53 deletion resulted in overexpression of NG2 (dotted area). (d) In the adjacent normal brain tissue of NSG SCID mouse injected with PDGFB/H3.3.K27M/p53−/− cells, NG2 overexpression was not detected. Insets are 40 × magnifications of the corresponding panel. Scale bar: 200 μM.
Figure 3. NG2 upregulation in DIPG is…
Figure 3. NG2 upregulation in DIPG is partially due to hypermethylation and downregulation of its regulatory microRNA, miR129-2
(a) Luciferase vector with 3′-UTR sequence of NG2 cloned downstream of the firefly luciferase gene (Luc-3′UTR) and plasmid vector with miR129-2 sequence were used. PDGFB mouse neurospheres were transfected in triplicates with luciferase and miR129-2 vectors, as shown, and luciferase expression was determined. Co-expression of Luc-3′UTR of NG2 and miR129-2 resulted in significant downregulation of luciferase expression as compared to cells transfected with luc-3′UTR and empty vector. P < 0.05 was considered significant. (b) PDGFB mouse neurospheres were transfected with miR129-2 or NG2-shRNA plasmids. Upregulation of either miR129-2 or NG2-shRNA resulted in NG2 downregulation in vitro when compared to empty vector or control shRNA treated cells. (c) Human DIPG cells, SF8628, were transduced with GFP expressing pCDH-control or pCDH-miR129-2 lentivirus. Immunofluorescence analysis by confocal microscopy revealed upregulation of miR129-2 (indicated by GFP) resulted in NG2 (red) downregulation in vitro when compared to control transduced cells. DAPI was used to stain nuclei. Scale bar = 5 μm. (d) Average expression of miR129-2 as assessed by RT-PCR (*miR129) or illumina expression chip assay (miR129), miR129 hypermethylation (miR129 me), and NG2 mRNA levels in human DIPG compared to adjacent normal. miR129-2 is shown to be downregulated in DIPG as assessed by RT-PCR and illumina chip assays. Hypermethylation of miR129-2 at four or more CpG sites corresponding to miR129-2 promoter were detected in 57% of DIPGs. As expected, downregulation of miR129-2 due to hypermethylation resulted in upregulation of NG2 mRNA as detected by NG2 mRNA using illumina platform. (e) To reverse miR129-2 hypermethylation, PDGFB mouse neurospheres were treated with 5-Azacytidine, a DNA methyl-transferase inhibitor drug. NG2 expression decreased in mouse tumor cells when treated with 10-30 μM 5-Azacytidine as compared to untreated cells. NG2 expression in each sample was normalized to corresponding GAPDH signal and average NG2 expression from three experiments was plotted.
Figure 3. NG2 upregulation in DIPG is…
Figure 3. NG2 upregulation in DIPG is partially due to hypermethylation and downregulation of its regulatory microRNA, miR129-2
(a) Luciferase vector with 3′-UTR sequence of NG2 cloned downstream of the firefly luciferase gene (Luc-3′UTR) and plasmid vector with miR129-2 sequence were used. PDGFB mouse neurospheres were transfected in triplicates with luciferase and miR129-2 vectors, as shown, and luciferase expression was determined. Co-expression of Luc-3′UTR of NG2 and miR129-2 resulted in significant downregulation of luciferase expression as compared to cells transfected with luc-3′UTR and empty vector. P < 0.05 was considered significant. (b) PDGFB mouse neurospheres were transfected with miR129-2 or NG2-shRNA plasmids. Upregulation of either miR129-2 or NG2-shRNA resulted in NG2 downregulation in vitro when compared to empty vector or control shRNA treated cells. (c) Human DIPG cells, SF8628, were transduced with GFP expressing pCDH-control or pCDH-miR129-2 lentivirus. Immunofluorescence analysis by confocal microscopy revealed upregulation of miR129-2 (indicated by GFP) resulted in NG2 (red) downregulation in vitro when compared to control transduced cells. DAPI was used to stain nuclei. Scale bar = 5 μm. (d) Average expression of miR129-2 as assessed by RT-PCR (*miR129) or illumina expression chip assay (miR129), miR129 hypermethylation (miR129 me), and NG2 mRNA levels in human DIPG compared to adjacent normal. miR129-2 is shown to be downregulated in DIPG as assessed by RT-PCR and illumina chip assays. Hypermethylation of miR129-2 at four or more CpG sites corresponding to miR129-2 promoter were detected in 57% of DIPGs. As expected, downregulation of miR129-2 due to hypermethylation resulted in upregulation of NG2 mRNA as detected by NG2 mRNA using illumina platform. (e) To reverse miR129-2 hypermethylation, PDGFB mouse neurospheres were treated with 5-Azacytidine, a DNA methyl-transferase inhibitor drug. NG2 expression decreased in mouse tumor cells when treated with 10-30 μM 5-Azacytidine as compared to untreated cells. NG2 expression in each sample was normalized to corresponding GAPDH signal and average NG2 expression from three experiments was plotted.
Figure 4. Mouse and human DIPG neurospheres…
Figure 4. Mouse and human DIPG neurospheres exhibit symmetric NG2 expression in vitro and are immature stem cells co-expressing oligodendrocyte (Olig2) and astrocyte (GFAP) markers
(a) PDGFB mouse tumor neurospheres (Mouse) and human primary DIPG cells (SF8628 and SUDIPGVI) were grown and stained using NG2 antibody (red) and DAPI (blue) nuclear staining. NG2 is equally expressed in dividing cells suggesting that its expression is defective (symmetric) in mitotic cells. Scale bar = 5 μm. (b) PDGFB mouse tumor (Mouse) and SF8628 human DIPG primary cells were expanded in culture and immunostained for DAPI (nuclear), oligodendrocyte marker (Olig2, green), NG2 (red), and astrocyte marker (GFAP, magenta). Confocal microscopy was used to assess expression of above-mentioned proteins in mitotic cells. Scale bar = 5 μM.
Figure 4. Mouse and human DIPG neurospheres…
Figure 4. Mouse and human DIPG neurospheres exhibit symmetric NG2 expression in vitro and are immature stem cells co-expressing oligodendrocyte (Olig2) and astrocyte (GFAP) markers
(a) PDGFB mouse tumor neurospheres (Mouse) and human primary DIPG cells (SF8628 and SUDIPGVI) were grown and stained using NG2 antibody (red) and DAPI (blue) nuclear staining. NG2 is equally expressed in dividing cells suggesting that its expression is defective (symmetric) in mitotic cells. Scale bar = 5 μm. (b) PDGFB mouse tumor (Mouse) and SF8628 human DIPG primary cells were expanded in culture and immunostained for DAPI (nuclear), oligodendrocyte marker (Olig2, green), NG2 (red), and astrocyte marker (GFAP, magenta). Confocal microscopy was used to assess expression of above-mentioned proteins in mitotic cells. Scale bar = 5 μM.
Figure 5. Orthotopic injection of NG2 expressing…
Figure 5. Orthotopic injection of NG2 expressing cells causes aggressive brainstem tumors in vivo resulting in a robust murine model of DIPG
(a) NG2 expressing neurospheres were isolated from PDGFB mouse pontine tumor and expanded in culture and stained for NG2 (red) and DAPI (blue) (b and c). Neurospheres were then injected into the brainstems of two-day-old (P2) mice. Whole brain was obtained at 3 weeks post injection and subjected to H&E staining (d-g). Injected cells show an infiltrative pattern of growth (g, arrow) at the site of injection. H&E staining also showed a large infiltrating tumor within pons (region p in d). 10 × magnifications (e-f) revealed the highly cellular tumor within the pons (e) that exhibits characteristics of DIPGs including tumor vascularity (f, arrows). Injected cells were infiltrated through the pons and away from the injection site (g, arrow). This pontine tumor is positive for the astrocytic marker GFAP (h), proliferation marker Ki67 (i), PDGFRβ (j), PDGFRα (k), and NG2 (l). Scale bar = 100 μM.
Figure 6. miR129-2 targets and downregulates NG2…
Figure 6. miR129-2 targets and downregulates NG2 in vivo
Brain sections from NG2-dsRed mice were analyzed 2 weeks after the intracranial injections with control lentiviral vector on left side and lentiviral vector harboring miR129-2 on right side. Detection of the green signal (GFP) indicates in vivo transduction of either control or miR129-2. Area enclosed by dotted line in 10 × image (Scale bar: 100 μM) represents the site of injection. 40 × magnification images (Scale bar: 10 μM) of boxed area in merged images are shown to demonstrate the downregulation of NG2 (red) in miR129-2 (green) transduced cells (arrows) while control transduced cells are expressing normal levels of NG2.

References

    1. Kohler BA, Ward E, McCarthy BJ, Schymura MJ, Ries LA, Eheman C, Jemal A, Anderson RN, Ajani UA, Edwards BK. Annual report to the nation on the status of cancer, 1975-2007, featuring tumors of the brain and other nervous system. J Natl Cancer Inst. 2011;103:714–736.
    1. Donaldson SS, Laningham F, Fisher PG. Advances toward an understanding of brainstem gliomas. J Clin Oncol. 2006;24:1266–1272.
    1. Jallo GI, Volkov A, Wong C, Carson BS, Sr, Penno MB. A novel brainstem tumor model: functional and histopathological characterization. Childs Nerv Syst. 2006;22:1519–1525.
    1. Rubin G, Michowitz S, Horev G, Herscovici Z, Cohen IJ, Shuper A, Rappaport ZH. Pediatric brain stem gliomas: an update. Childs Nerv Syst. 1998;14:167–173.
    1. Khuong-Quang DA, Buczkowicz P, Rakopoulos P, Liu XY, Fontebasso AM, Bouffet E, Bartels U, Albrecht S, Schwartzentruber J, Letourneau L, Bourgey M, Bourque G, Montpetit A, Bourret G, Lepage P, Fleming A, et al. K27M mutation in histone H3. 3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta neuropathologica. 2012;124:439–447.
    1. Puget S, Philippe C, Bax DA, Job B, Varlet P, Junier MP, Andreiuolo F, Carvalho D, Reis R, Guerrini-Rousseau L, Roujeau T, Dessen P, Richon C, Lazar V, Le Teuff G, Sainte-Rose C, et al. Mesenchymal transition and PDGFRA amplification/mutation are key distinct oncogenic events in pediatric diffuse intrinsic pontine gliomas. PloS one. 2012;7:e30313.
    1. Monje M, Mitra SS, Freret ME, Raveh TB, Kim J, Masek M, Attema JL, Li G, Haddix T, Edwards MS, Fisher PG, Weissman IL, Rowitch DH, Vogel H, Wong AJ, Beachy PA. Hedgehog-responsive candidate cell of origin for diffuse intrinsic pontine glioma. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:4453–4458.
    1. Saratsis A, Kambhampati M, Snyder K, Yadavilli S, Devaney J, Harmon B, Hall J, Raabe E, An P, Weingart M, Rood B, Magge S, MacDonald T, Packer R, Nazarian J. Comparative multidimensional molecular analyses of pediatric diffuse intrinsic pontine glioma reveals distinct molecular subtypes. Acta neuropathologica. 2014;127:881–895.
    1. Price MA, Colvin Wanshura LE, Yang J, Carlson J, Xiang B, Li G, Ferrone S, Dudek AZ, Turley EA, McCarthy JB. CSPG4, a potential therapeutic target, facilitates malignant progression of melanoma. Pigment Cell Melanoma Res. 2011;24:1148–1157.
    1. Chekenya M, Enger PO, Thorsen F, Tysnes BB, Al-Sarraj S, Read TA, Furmanek T, Mahesparan R, Levine JM, Butt AM, Pilkington GJ, Bjerkvig R. The glial precursor proteoglycan, NG2, is expressed on tumour neovasculature by vascular pericytes in human malignant brain tumours. Neuropathol Appl Neurobiol. 2002;28:367–380.
    1. Chekenya M, Rooprai HK, Davies D, Levine JM, Butt AM, Pilkington GJ. The NG2 chondroitin sulfate proteoglycan: role in malignant progression of human brain tumours. Int J Dev Neurosci. 1999;17:421–435.
    1. Ligon KL, Alberta JA, Kho AT, Weiss J, Kwaan MR, Nutt CL, Louis DN, Stiles CD, Rowitch DH. The oligodendroglial lineage marker OLIG2 is universally expressed in diffuse gliomas. J Neuropathol Exp Neurol. 2004;63:499–509.
    1. Sugiarto S, Persson AI, Munoz EG, Waldhuber M, Lamagna C, Andor N, Hanecker P, Ayers-Ringler J, Phillips J, Siu J, Lim DA, Vandenberg S, Stallcup W, Berger MS, Bergers G, Weiss WA, et al. Asymmetry-defective oligodendrocyte progenitors are glioma precursors. Cancer Cell. 2011;20:328–340.
    1. Gielen GH, Gessi M, Hammes J, Kramm CM, Waha A, Pietsch T. H3F3A K27M mutation in pediatric CNS tumors: a marker for diffuse high-grade astrocytomas. American journal of clinical pathology. 2013;139:345–349.
    1. Wu G, Broniscer A, McEachron TA, Lu C, Paugh BS, Becksfort J, Qu C, Ding L, Huether R, Parker M, Zhang J, Gajjar A, Dyer MA, Mullighan CG, Gilbertson RJ, Mardis ER, et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet. 2012;44:251–253.
    1. Becher OJ, Hambardzumyan D, Walker TR, Helmy K, Nazarian J, Albrecht S, Hiner RL, Gall S, Huse JT, Jabado N, MacDonald TJ, Holland EC. Preclinical evaluation of radiation and perifosine in a genetically and histologically accurate model of brainstem glioma. Cancer research. 2010;70:2548–2557.
    1. Lewis PW, Muller MM, Koletsky MS, Cordero F, Lin S, Banaszynski LA, Garcia BA, Muir TW, Becher OJ, Allis CD. Inhibition of PRC2 Activity by a Gain-of-Function H3 Mutation Found in Pediatric Glioblastoma. Science. 2013
    1. Tsai KW, Wu CW, Hu LY, Li SC, Liao YL, Lai CH, Kao HW, Fang WL, Huang KH, Chan WC, Lin WC. Epigenetic regulation of miR-34b and miR-129 expression in gastric cancer. International journal of cancer Journal international du cancer. 2011;129:2600–2610.
    1. Alonso MM, Diez-Valle R, Manterola L, Rubio A, Liu D, Cortes-Santiago N, Urquiza L, Jauregi P, Lopez de Munain A, Sampron N, Aramburu A, Tejada-Solis S, Vicente C, Odero MD, Bandres E, Garcia-Foncillas J, et al. Genetic and epigenetic modifications of Sox2 contribute to the invasive phenotype of malignant gliomas. PloS one. 2011;6:e26740.
    1. Cory G. Scratch-wound assay. Methods Mol Biol. 769:25–30.
    1. Broniscer A, Gajjar A. Supratentorial high-grade astrocytoma and diffuse brainstem glioma: two challenges for the pediatric oncologist. Oncologist. 2004;9:197–206.
    1. Jansen MH, van Vuurden DG, Vandertop WP, Kaspers GJ. Diffuse intrinsic pontine gliomas: a systematic update on clinical trials and biology. Cancer Treat Rev. 2011;38:27–35.
    1. Misuraca KL, Barton KL, Chung A, Diaz AK, Conway SJ, Corcoran DL, Baker SJ, Becher OJ. Pax3 expression enhances PDGF-B-induced brainstem gliomagenesis and characterizes a subset of brainstem glioma. Acta Neuropathol Commun. 2014;2:134.
    1. Zhu X, Hill RA, Nishiyama A. NG2 cells generate oligodendrocytes and gray matter astrocytes in the spinal cord. Neuron Glia Biol. 2008;4:19–26.
    1. Burg MA, Grako KA, Stallcup WB. Expression of the NG2 proteoglycan enhances the growth and metastatic properties of melanoma cells. J Cell Physiol. 1998;177:299–312.
    1. Schrappe M, Klier FG, Spiro RC, Waltz TA, Reisfeld RA, Gladson CL. Correlation of chondroitin sulfate proteoglycan expression on proliferating brain capillary endothelial cells with the malignant phenotype of astroglial cells. Cancer research. 1991;51:4986–4993.
    1. Shoshan Y, Nishiyama A, Chang A, Mork S, Barnett GH, Cowell JK, Trapp BD, Staugaitis SM. Expression of oligodendrocyte progenitor cell antigens by gliomas: implications for the histogenesis of brain tumors. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:10361–10366.
    1. Wiranowska M, Ladd S, Smith SR, Gottschall PE. CD44 adhesion molecule and neuro-glial proteoglycan NG2 as invasive markers of glioma. Brain Cell Biol. 2006;35:159–172.
    1. Chekenya M, Krakstad C, Svendsen A, Netland IA, Staalesen V, Tysnes BB, Selheim F, Wang J, Sakariassen PO, Sandal T, Lonning PE, Flatmark T, Enger PO, Bjerkvig R, Sioud M, Stallcup WB. The progenitor cell marker NG2/MPG promotes chemoresistance by activation of integrin-dependent PI3K/Akt signaling. Oncogene. 2008;27:5182–5194.
    1. Burg MA, Tillet E, Timpl R, Stallcup WB. Binding of the NG2 proteoglycan to type VI collagen and other extracellular matrix molecules. The Journal of biological chemistry. 1996;271:26110–26116.
    1. Dawson MR, Polito A, Levine JM, Reynolds R. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci. 2003;24:476–488.
    1. Nishiyama A, Komitova M, Suzuki R, Zhu X. Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat Rev Neurosci. 2009;10:9–22.
    1. Zhu X, Hill RA, Dietrich D, Komitova M, Suzuki R, Nishiyama A. Age-dependent fate and lineage restriction of single NG2 cells. Development. 2011;138:745–753.
    1. Glasgow SM, Zhu W, Stolt CC, Huang TW, Chen F, LoTurco JJ, Neul JL, Wegner M, Mohila C, Deneen B. Mutual antagonism between Sox10 and NFIA regulates diversification of glial lineages and glioma subtypes. Nature neuroscience. 2014;17:1322–1329.
    1. Liu C, Sage JC, Miller MR, Verhaak RG, Hippenmeyer S, Vogel H, Foreman O, Bronson RT, Nishiyama A, Luo L, Zong H. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell. 2011;146:209–221.
    1. Tasic B, Hippenmeyer S, Wang C, Gamboa M, Zong H, Chen-Tsai Y, Luo L. Site-specific integrase-mediated transgenesis in mice via pronuclear injection. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:7902–7907.
    1. Aoki Y, Hashizume R, Ozawa T, Banerjee A, Prados M, James CD, Gupta N. An experimental xenograft mouse model of diffuse pontine glioma designed for therapeutic testing. J Neurooncol. 2012;108:29–35.
    1. Kambhampati M, Perez JP, Yadavilli S, Saratsis AM, Hill AD, Ho C-Y, Panditharatna E, Markel M, Packer RJ, Nazarian J. A standardized autopsy procurement allows for the comprehensive study of DIPG biology. Oncotarget. 2015 (epub ahead of print)
    1. Hashizume R, Smirnov I, Liu S, Phillips JJ, Hyer J, McKnight TR, Wendland M, Prados M, Banerjee A, Nicolaides T, Mueller S, James CD, Gupta N. Characterization of a diffuse intrinsic pontine glioma cell line: implications for future investigations and treatment. Journal of neuro-oncology. 2012;110:305–313.
    1. Yue PY, Leung EP, Mak NK, Wong RN. A simplified method for quantifying cell migration/wound healing in 96-well plates. J Biomol Screen. 2010;15:427–433.

Source: PubMed

3
Abonnere