Tumour compartment transcriptomics demonstrates the activation of inflammatory and odontogenic programmes in human adamantinomatous craniopharyngioma and identifies the MAPK/ERK pathway as a novel therapeutic target

John R Apps, Gabriela Carreno, Jose Mario Gonzalez-Meljem, Scott Haston, Romain Guiho, Julie E Cooper, Saba Manshaei, Nital Jani, Annett Hölsken, Benedetta Pettorini, Robert J Beynon, Deborah M Simpson, Helen C Fraser, Ying Hong, Shirleen Hallang, Thomas J Stone, Alex Virasami, Andrew M Donson, David Jones, Kristian Aquilina, Helen Spoudeas, Abhijit R Joshi, Richard Grundy, Lisa C D Storer, Márta Korbonits, David A Hilton, Kyoko Tossell, Selvam Thavaraj, Mark A Ungless, Jesus Gil, Rolf Buslei, Todd Hankinson, Darren Hargrave, Colin Goding, Cynthia L Andoniadou, Paul Brogan, Thomas S Jacques, Hywel J Williams, Juan Pedro Martinez-Barbera, John R Apps, Gabriela Carreno, Jose Mario Gonzalez-Meljem, Scott Haston, Romain Guiho, Julie E Cooper, Saba Manshaei, Nital Jani, Annett Hölsken, Benedetta Pettorini, Robert J Beynon, Deborah M Simpson, Helen C Fraser, Ying Hong, Shirleen Hallang, Thomas J Stone, Alex Virasami, Andrew M Donson, David Jones, Kristian Aquilina, Helen Spoudeas, Abhijit R Joshi, Richard Grundy, Lisa C D Storer, Márta Korbonits, David A Hilton, Kyoko Tossell, Selvam Thavaraj, Mark A Ungless, Jesus Gil, Rolf Buslei, Todd Hankinson, Darren Hargrave, Colin Goding, Cynthia L Andoniadou, Paul Brogan, Thomas S Jacques, Hywel J Williams, Juan Pedro Martinez-Barbera

Abstract

Adamantinomatous craniopharyngiomas (ACPs) are clinically challenging tumours, the majority of which have activating mutations in CTNNB1. They are histologically complex, showing cystic and solid components, the latter comprised of different morphological cell types (e.g. β-catenin-accumulating cluster cells and palisading epithelium), surrounded by a florid glial reaction with immune cells. Here, we have carried out RNA sequencing on 18 ACP samples and integrated these data with an existing ACP transcriptomic dataset. No studies so far have examined the patterns of gene expression within the different cellular compartments of the tumour. To achieve this goal, we have combined laser capture microdissection with computational analyses to reveal groups of genes that are associated with either epithelial tumour cells (clusters and palisading epithelium), glial tissue or immune infiltrate. We use these human ACP molecular signatures and RNA-Seq data from two ACP mouse models to reveal that cell clusters are molecularly analogous to the enamel knot, a critical signalling centre controlling normal tooth morphogenesis. Supporting this finding, we show that human cluster cells express high levels of several members of the FGF, TGFB and BMP families of secreted factors, which signal to neighbouring cells as evidenced by immunostaining against the phosphorylated proteins pERK1/2, pSMAD3 and pSMAD1/5/9 in both human and mouse ACP. We reveal that inhibiting the MAPK/ERK pathway with trametinib, a clinically approved MEK inhibitor, results in reduced proliferation and increased apoptosis in explant cultures of human and mouse ACP. Finally, we analyse a prominent molecular signature in the glial reactive tissue to characterise the inflammatory microenvironment and uncover the activation of inflammasomes in human ACP. We validate these results by immunostaining against immune cell markers, cytokine ELISA and proteome analysis in both solid tumour and cystic fluid from ACP patients. Our data support a new molecular paradigm for understanding ACP tumorigenesis as an aberrant mimic of natural tooth development and opens new therapeutic opportunities by revealing the activation of the MAPK/ERK and inflammasome pathways in human ACP.

Keywords: Craniopharyngioma; IL1-β; Inflammasome; MAPK/ERK pathway; Odontogenesis; Paracrine signalling; Trametinib.

Conflict of interest statement

P.B. has received institutional grants from SOBI, Roche and Novartis; consultancy fees from Roche; and lecturing fees from SOBI and Novartis. The other authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
Bioinformatics analysis of gene expression profiling of human ACP whole tumours identifies modules of genes potentially associated with specific tumour cell compartments. a Representative histology of ACP samples showing and areas of tumour (T), reactive glial tissue (G), wet keratin/ghost cells (WK), epithelial whorls (C) (epithelial whorls referred to in this paper as clusters), palisading epithelium (PE) and stellate reticulum (SR). Immunohistochemistry using antibodies against β-catenin (β-cat) on case JA029 showing clusters of cells with nuclear-cytoplasmic accumulation. Scale bars 400 μm (top left panel), 100 μm (other three panels). b Scatterplot revealing a significant correlation between CTNNB1 mutation frequency and histologically assessed tumour content. # case JA011; * case JA005; controls: fetal pituitaries and NFPA tissues. See Suppl. Table 1 (Online Resource 2) for sample details. c Principal component analysis plot showing the separation between adamantinomatous craniopharyngioma (ACP), non-functioning pituitary adenoma (NFPA) and control fetal pituitary tissues (fetal). d Bar plot of selected statistically significant and differentially expressed genes, as assessed by DESeq2, in ACP tumours compared with control fetal tissue. Higher than 0 means higher expression in ACP and lower than 0 means higher in control tissue. The most up-regulated genes in ACP tumours are keratins. Other up-regulated genes include WNTs and WNT pathway targets (e.g. NOTUM, AXIN2, LEF1), genes known to be expressed in ACP (e.g. FGFs, BMPs) and previously suggested therapeutic targets (SHH, MMP12, MMP9, EGFR). Pituitary transcription factors (LHX3, POU1F1) and pituitary hormones (e.g. FSHB, GH1, TSHB) are up-regulated in controls. See Suppl. Table 3 (Online Resource 5) for details. Error bars = 1 standard error, *** adjusted p value < 1 × 10−9. e Multidimensional scaling plot of expression patterns of the 5000 most differentially expressed genes included in the weighted gene co-expression network analysis (WGCNA) analysis. The colour of each gene indicates it membership to a co-expressed gene expression module. f Heatmap of correlations between each module’s gene expression profile and phenotypic information. Scale bar indicate r value − 1 to + 1. For instance, the brown module shows a strong correlation with tumour content and mutational frequency, whilst the blue module correlates with the presence of glial reactive tissue and GFAP
Fig. 2
Fig. 2
Gene profiling of laser capture microdissected tumour cells confirms the WGCNA, identifying modules associated with tumour cell compartments and revealing novel ACP genes. a Scheme of the experimental approach. Histological sections of tumour samples JA004 and JA029 were subjected to laser capture microdissection (LCM) to isolate β-catenin-accumulating cell clusters (C), palisading epithelium (PE) and glial reactive tissue (G). Biological duplicates were performed for clusters and palisading epithelium in case JA004. RNA was purified from each of these tumour cell compartments, amplified and sequenced. b Principal component analysis reveals grouping of the data from laser capture microdissected samples. c Gene set enrichment analysis revealing the enrichment of an inflammatory signature in microdissected glial reactive tissue relative to tumour tissue (i.e. genes included in clusters plus palisading epithelium), whilst a WNT signalling expression signature is associated with the microdissected tumour tissue. Enrichment for WNT signalling was stronger in the clusters relative to both PE and glial tissue. d Gene set enrichment analysis showing the enrichment of the brown module genes with a signature of tumour cell compartments (i.e. genes including in cluster cells plus palisading epithelium). In contrast, both the blue and turquoise module genes are predominantly expressed by reactive glia. e Double immunofluorescence staining revealing the expression of BCL11B and TP63 in the epithelial components of the tumour, including palisading epithelium (PE) and β-catenin accumulating clusters (C), but not in reactive glial tissue (G). NES normalised enrichment score, FDR false discovery rate. Scale bars 100 μm
Fig. 3
Fig. 3
ACP and developing mammalian tooth share common molecular signatures. a Gene set enrichment plots showing that ACP tumours are enriched for genes expressed by ameloblasts and inner enamel epithelium. Results obtained from using the RNA dataset from profiling whole ACP tumours. b Expression of relevant ameloblast-related genes is significantly expressed at higher levels in whole ACP tumours compared with control tissues (fetal pituitary and NFPA). See Suppl. Table 5 (Online Resource 7) for details (*** adjusted p value < 1 × 10−7, * adjusted p value = 0.028). c Gene set enrichment plots showing that the cluster cells are enriched for genes expressed in the enamel knot whilst palisading epithelium shows a signature of inner enamel epithelia at cap stage. Results obtained from using the RNA dataset from profiling microdissected ACP compartments. d Double immunofluorescent staining reveals the co-expression of p21/CDKN1A and EDAR, two enamel knot markers, in the β-catenin-accumulating clusters. e Gene set enrichment analysis showing that both the inner enamel epithelium and ameloblast gene signatures are enriched in the embryonic mouse ACP model compared with wild-type controls (WT). f Enrichment plots confirming that mouse clusters from both the embryonic and inducible ACP mouse models show a molecular signature of the enamel knot. NES normalised enrichment score, FDR false discovery rate. Scale bars 100 μm
Fig. 4
Fig. 4
Identification of the activation of the MAPK/ERK, TGFB and BMP signalling pathways in human ACP. a Immunohistochemistry revealing the expression of phosphorylated ERK1/2 (pERK1/2), a read out of active MAPK/ERK pathway, at the tips of the invading tumour epithelium (palisading epithelium, arrows in a, a‴ and a″″) and within reactive glial tissue (G; arrows in a″). b Double immunofluorescent staining showing pERK1/2 expression in the palisading epithelium (PE) around the β-catenin accumulating clusters (C), which express several activating ligands of the MAPK/ERK pathway (see main text for details). Note that cells within the reactive glial tissue (G) are also pERK1/2 positive. c Double immunofluorescence revealing abundant Ki67+ve cells in the palisading epithelium close to clusters. d Double immunofluorescence showing Ki67 and pERK1/2 co-expression within the palisading epithelium (PE). e Double immunofluorescence showing pSMAD3 staining, indicating activation of TGFβ signalling, in both tumour and reactive glia, with strongest signal in reactive tissue adjacent to tumour epithelia (arrowhead). Double immunofluorescence reveals pSMAD1/5/9 staining, indicating BMP signalling in cells within and adjacent to the β-catenin-accumulating clusters (C). Note the absence of staining in the palisading epithelium (PE). Scale bars: aa″″ 200 μm; bf 100 μm
Fig. 5
Fig. 5
Identification of the activation of the MAPK/ERK, TGFB and BMP signalling pathways in the ACP embryonic mouse model. Double immunofluorescent staining on histological sections of neoplastic pituitaries of the ACP embryonic mouse model at postnatal day 1 (P1). Note the widespread expression of pERK1/2 in cells around the β-catenin-accumulating cell clusters, which show no expression of this marker. pSMAD3 and pSMAD1/5/9 staining is also predominant in cells surrounding the clusters, but occasionally weak staining is observed in some cluster cells (arrowheads). Scale bars 50 μm
Fig. 6
Fig. 6
Ex vivo inhibition of the MAPK/ERK pathway in mouse ACP results in decreased proliferation and increased apoptosis of tumour cells. Neoplastic pituitaries of the ACP embryonic mouse model were cultured in the presence of the MEK inhibitor trametinib (2 or 20 nM) or the vehicle control (DMSO) for 18 h. Following histological processing, sections were immunostained against β-catenin and pERK1/2 (readout of active MAPK/ERK pathway; a), Ki-67 (proliferation marker; b) and cleaved caspase-3 (apoptosis marker; c). Quantitative analysis showing a significant dose-dependent reduction in Ki-67 proliferative index (d; 20 nM) and an increase in apoptosis (e; 2 and 20 nM) in trametinib-treated relative to vehicle-treated control. Kruskal–Wallis with Dunn’s post-test **p < 0.01; ***p < 0.001. Mean of 4.1 × 103 nuclei for each point. Scale bars 50 μm
Fig. 7
Fig. 7
Ex vivo inhibition of the MAPK/ERK pathway in human ACP results in decreased proliferation and increased apoptosis of tumour cells. Small pieces of three human ACP tumours were cultured in the presence of the MEK inhibitor trametinib (2 or 20 nM) or the vehicle control (DMSO) for 18 h. Following histological processing, sections were immunostained against β-catenin and pERK1/2 (readout of active MAPK/ERK pathway; a), Ki-67 (proliferation marker; b) and cleaved caspase-3 (apoptosis marker; c). Quantitative analysis showing a significant dose-dependent reduction in Ki-67 proliferative index (d; 2 and 20 nM) and an increase in apoptosis (e; 20 nM) in trametinib-treated relative to vehicle-treated control. Kruskal–Wallis with Dunn’s post-test **p < 0.01; ***p < 0.001. Mean of 1.6 × 104 nuclei for each point. Scale bars 50 μm
Fig. 8
Fig. 8
Characterisation of the immune microenvironment in the solid component and cystic fluid of human ACP. a Immunohistochemistry showing infiltration of myeloid (CD68+ve) and lymphoid (CD3+ve) within human ACP tumour (T) and reactive glial tissue (G). CD68+ve and IBA1+ve immunostaining is observed in close association with the cholesterol clefts (arrows). Likewise, immunohistochemistry against the chemokine CCL2 is detected near the cholesterol clefts. b The expression of the cytokines IL18, IL1B and IL10 correlate significantly with CD14 expression, a marker preferentially expressed in monocytes/macrophages, in the 24 samples (ACP tumours and control tissues) profiled by RNA-Seq. c Multiplex ELISA quantification of cytokine protein expression within the solid tumour (n = 8 tumours; left) and cystic fluid (n = 10 samples; right). IL8, IL18, IL6 and IL1B are the highest expressed cytokines in the solid tumour. In the cystic fluid, levels of IL6 and IL8 are the highest, but all the other cytokines are also detected. For solid tumour values were normalised against total protein and for cystic fluid samples against volume. The blue dots represent the value obtained of each cytokine for each sample and the red dots represent the median. Scale bars 100 μm
Fig. 9
Fig. 9
Activation of the inflammasomes underlies the ACP inflammatory response. a Gene set enrichment plots showing that human ACP tumours are enriched for genes expressed by macrophages, chondrocytes and uterine muscle cells exposed to IL1B in culture conditions. b Gene set enrichment plots reveal a molecular signature of atherosclerotic plaques in human ACP. Results are based on data obtained from RNA-Seq profiles from whole ACP tumours and control tissues (fetal pituitaries and NFPA). c The levels of IL6, IL8 and TNFα protein correlate with levels of IL1B in human ACP cystic fluid (n = 10 cystic fluid samples; determined by ELISA). NES normalised enrichment score, FDR false discovery rate
Fig. 10
Fig. 10
Schematic summary of the findings. Molecular and histological relationships between ACP pathogenesis and tooth development. The enamel knot and the β-catenin-accumulating clusters, which both have similar expression profiles and comparable histology, act as signalling hubs through the secretion of a several growth factors acting in an autocrine and/or paracrine manner on the surrounding cells, i.e. the enamel epithelium/dental mesenchyme in the forming tooth or the palisading epithelium, stellate reticulum and glial reactive tissue in ACP. Reciprocal signalling from surrounding tissues to the enamel knot and clusters is indicated by double-headed arrows. In the glial reactive tissue, cholesterol crystals activate the inflammasomes resulting in the secretion of IL1B, which in turn acts on the local immune effector cells to drive an inflammatory response

References

    1. Andoniadou CL, Gaston-Massuet C, Reddy R, Schneider RP, Blasco MA, Le Tissier P, Jacques TS, Pevny LH, Dattani MT, Martinez-Barbera JP. Identification of novel pathways involved in the pathogenesis of human adamantinomatous craniopharyngioma. Acta Neuropathol. 2012;124:259–271. doi: 10.1007/s00401-012-0957-9.
    1. Andoniadou CL, Matsushima D, Mousavy Gharavy SN, Signore M, Mackintosh AI, Schaeffer M, Gaston-Massuet C, Mollard P, Jacques TS, Le Tissier P, et al. Sox2(+) stem/progenitor cells in the adult mouse pituitary support organ homeostasis and have tumor-inducing potential. Cell Stem Cell. 2013;13:433–445. doi: 10.1016/j.stem.2013.07.004.
    1. Apps JR, Hutchinson JC, Arthurs OJ, Virasami A, Joshi A, Zeller-Plumhoff B, Moulding D, Jacques TS, Sebire NJ, Martinez-Barbera JP. Imaging invasion: micro-CT imaging of adamantinomatous craniopharyngioma highlights cell type specific spatial relationships of tissue invasion. Acta Neuropathol Commun. 2016;4:57. doi: 10.1186/s40478-016-0321-8.
    1. Apps JR, Martinez-Barbera JP. Genetically engineered mouse models of craniopharyngioma: an opportunity for therapy development and understanding of tumor biology. Brain Pathol. 2017;27:364–369. doi: 10.1111/bpa.12501.
    1. Apte RN, Voronov E. Is interleukin-1 a good or bad ‘guy’ in tumor immunobiology and immunotherapy? Immunol Rev. 2008;222:222–241. doi: 10.1111/j.1600-065X.2008.00615.x.
    1. Beaty NB, Ahn E. Images in clinical medicine. Adamantinomatous craniopharyngioma containing teeth. N Engl J Med. 2014;370:860. doi: 10.1056/NEJMicm1308260.
    1. Bernstein ML, Buchino JJ. The histologic similarity between craniopharyngioma and odontogenic lesions: a reappraisal. Oral Surg Oral Med Oral Pathol. 1983;56:502–511. doi: 10.1016/0030-4220(83)90098-1.
    1. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-Santagata D, Thorner AR, Lawrence MS, Rodriguez FJ, Bernardo LA, Schubert L, et al. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nat Genet. 2014;46:161–165. doi: 10.1038/ng.2868.
    1. Burghaus S, Holsken A, Buchfelder M, Fahlbusch R, Riederer BM, Hans V, Blumcke I, Buslei R. A tumor-specific cellular environment at the brain invasion border of adamantinomatous craniopharyngiomas. Virchows Arch. 2010;456:287–300. doi: 10.1007/s00428-009-0873-0.
    1. Buslei R, Holsken A, Hofmann B, Kreutzer J, Siebzehnrubl F, Hans V, Oppel F, Buchfelder M, Fahlbusch R, Blumcke I. Nuclear beta-catenin accumulation associates with epithelial morphogenesis in craniopharyngiomas. Acta Neuropathol. 2007;113:585–590. doi: 10.1007/s00401-006-0184-3.
    1. Cao J, Lin JP, Yang LX, Chen K, Huang ZS. Expression of aberrant beta-catenin and impaired p63 in craniopharyngiomas. Br J Neurosurg. 2010;24:249–256. doi: 10.3109/02688690903576237.
    1. Cavalcanti NG, Marques CD, Lins ELTU, Pereira MC, Rego MJ, Duarte AL, Pitta Ida R, Pitta MG. Cytokine profile in gout: inflammation driven by IL-6 and IL-18? Immunol Investig. 2016;45:383–395. doi: 10.3109/08820139.2016.1153651.
    1. Donson A, Apps JR, Greisinger AM, Amani V, Witt DA, Anderson RC, Niazi TN, Grant G, Souweidane M, Johnson JM, et al. Molecular analyses reveal inflammatory mediators in the solid component and cyst fluid of human adamantinomatous craniopharyngioma. J Neuropathol Exp Neurol. 2017
    1. Gaston-Massuet C, Andoniadou CL, Signore M, Jayakody SA, Charolidi N, Kyeyune R, Vernay B, Jacques TS, Taketo MM, Le Tissier P, et al. Increased wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proc Natl Acad Sci USA. 2011;108:11482–11487. doi: 10.1073/pnas.1101553108.
    1. Goldbach-Mansky R. Current status of understanding the pathogenesis and management of patients with NOMID/CINCA. Curr Rheumatol Rep. 2011;13:123–131. doi: 10.1007/s11926-011-0165-y.
    1. Gomes DC, Jamra SA, Leal LF, Colli LM, Campanini ML, Oliveira RS, Martinelli CE, Jr, Elias PC, Moreira AC, Machado HR, et al. Sonic Hedgehog pathway is upregulated in adamantinomatous craniopharyngiomas. Eur J Endocrinol. 2015;172:603–608. doi: 10.1530/EJE-14-0934.
    1. Gonzalez-Meljem JM, Haston S, Carreno G, Apps JR, Pozzi S, Stache C, Kaushal G, Virasami A, Panousopoulos L, Mousavy-Gharavy NS, et al. Stem cell senescence drives age-attenuated induction of pituitary tumours in mouse models of paediatric craniopharyngioma. Nat Commun. 2017;8:1819. doi: 10.1038/s41467-017-01992-5.
    1. Goodwin AF, Tidyman WE, Jheon AH, Sharir A, Zheng X, Charles C, Fagin JA, McMahon M, Diekwisch TG, Ganss B, et al. Abnormal Ras signaling in Costello syndrome (CS) negatively regulates enamel formation. Hum Mol Genet. 2014;23:682–692. doi: 10.1093/hmg/ddt455.
    1. Gorlin RJ, Chaudhry AP. The ameloblastoma and the craniopharyngioma; their similarities and differences. Oral Surg Oral Med Oral Pathol. 1959;12:199–205. doi: 10.1016/0030-4220(59)90144-6.
    1. Gritli-Linde A, Bei M, Maas R, Zhang XM, Linde A, McMahon AP. Shh signaling within the dental epithelium is necessary for cell proliferation, growth and polarization. Development. 2002;129:5323–5337. doi: 10.1242/dev.00100.
    1. Gump JM, Donson AM, Birks DK, Amani VM, Rao KK, Griesinger AM, Kleinschmidt-DeMasters BK, Johnston JM, Anderson RC, Rosenfeld A, et al. Identification of targets for rational pharmacological therapy in childhood craniopharyngioma. Acta Neuropathol Commun. 2015;3:30. doi: 10.1186/s40478-015-0211-5.
    1. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–687. doi: 10.1038/nm.3893.
    1. Hassanein AM, Glanz SM, Kessler HP, Eskin TA, Liu C. beta-Catenin is expressed aberrantly in tumors expressing shadow cells. Pilomatricoma, craniopharyngioma, and calcifying odontogenic cyst. Am J Clin Pathol. 2003;120:732–736. doi: 10.1309/EALEG7LD6W7167PX.
    1. Holsken A, Gebhardt M, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. EGFR signaling regulates tumor cell migration in craniopharyngiomas. Clin Cancer Res. 2011;17:4367–4377. doi: 10.1158/1078-0432.CCR-10-2811.
    1. Holsken A, Kreutzer J, Hofmann BM, Hans V, Oppel F, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. Target gene activation of the Wnt signaling pathway in nuclear beta-catenin accumulating cells of adamantinomatous craniopharyngiomas. Brain Pathol. 2009;19:357–364. doi: 10.1111/j.1750-3639.2008.00180.x.
    1. Holsken A, Sill M, Merkle J, Schweizer L, Buchfelder M, Flitsch J, Fahlbusch R, Metzler M, Kool M, Pfister SM, et al. Adamantinomatous and papillary craniopharyngiomas are characterized by distinct epigenomic as well as mutational and transcriptomic profiles. Acta Neuropathol Commun. 2016;4:20. doi: 10.1186/s40478-016-0287-6.
    1. Jarvinen E, Salazar-Ciudad I, Birchmeier W, Taketo MM, Jernvall J, Thesleff I. Continuous tooth generation in mouse is induced by activated epithelial Wnt/beta-catenin signaling. Proc Natl Acad Sci USA. 2006;103:18627–18632. doi: 10.1073/pnas.0607289103.
    1. Jussila M, Thesleff I. Signaling networks regulating tooth organogenesis and regeneration, and the specification of dental mesenchymal and epithelial cell lineages. Cold Spring Harb Perspect Biol. 2012;4:a008425. doi: 10.1101/cshperspect.a008425.
    1. Kalnins V. Calcification and amelogenesis in craniopharyngiomas. Oral Surg Oral Med Oral Pathol. 1971;31:366–379. doi: 10.1016/0030-4220(71)90159-9.
    1. Kaski M, Nieminen P, Salhlberg C, Aberg T, Thesleff I (1996–2007) Developmental biology programme of the University of Helsinki. Gene expression in tooth.
    1. Kato K, Nakatani Y, Kanno H, Inayama Y, Ijiri R, Nagahara N, Miyake T, Tanaka M, Ito Y, Aida N, et al. Possible linkage between specific histological structures and aberrant reactivation of the Wnt pathway in adamantinomatous craniopharyngioma. J Pathol. 2004;203:814–821. doi: 10.1002/path.1562.
    1. Kikuchi K, Ito S, Inoue H, Gonzalez-Alva P, Miyazaki Y, Sakashita H, Yoshino A, Katayama Y, Terui T, Ide F, et al. Immunohistochemical expression of podoplanin in so-called hard alpha-keratin-expressing tumors, including calcifying cystic odontogenic tumor, craniopharyngioma, and pilomatrixoma. J Oral Sci. 2012;54:165–175. doi: 10.2334/josnusd.54.165.
    1. Kingsbury SR, Conaghan PG, McDermott MF. The role of the NLRP3 inflammasome in gout. J Inflamm Res. 2011;4:39–49.
    1. Kinsler VA, O’Hare P, Jacques T, Hargrave D, Slater O. MEK inhibition appears to improve symptom control in primary NRAS-driven CNS melanoma in children. Br J Cancer. 2017;116:990–993. doi: 10.1038/bjc.2017.49.
    1. Larkin SJ, Ansorge O. Pathology and pathogenesis of craniopharyngiomas. Pituitary. 2013;16:9–17. doi: 10.1007/s11102-012-0418-4.
    1. Lee JJ, Perera RM, Wang H, Wu DC, Liu XS, Han S, Fitamant J, Jones PD, Ghanta KS, Kawano S, et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci USA. 2014;111:E3091–E3100. doi: 10.1073/pnas.1411679111.
    1. Lewis AM, Varghese S, Xu H, Alexander HR (2006) Interleukin-1 and cancer progression: the emerging role of interleukin-1 receptor antagonist as a novel therapeutic agent in cancer treatment. J Transl Med 4:48. 10.1186/1479-5876-4-48
    1. Liu F, Dangaria S, Andl T, Zhang Y, Wright AC, Damek-Poprawa M, Piccolo S, Nagy A, Taketo MM, Diekwisch TG, et al. beta-Catenin initiates tooth neogenesis in adult rodent incisors. J Dent Res. 2010;89:909–914. doi: 10.1177/0022034510370090.
    1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK. World Health Organisation histological classification of tumours of the central nervous system. Lyon: International Agency for Research on Cancer; 2016.
    1. Martinez-Barbera JP. Molecular and cellular pathogenesis of adamantinomatous craniopharyngioma. Neuropathol Appl Neurobiol. 2015
    1. Martinez-Barbera JP, Buslei R. Adamantinomatous craniopharyngioma: pathology, molecular genetics and mouse models. J Pediatr Endocrinol Metab. 2015;28:7–17. doi: 10.1515/jpem-2014-0442.
    1. Muller C, Adroos N, Lockhat Z, Slavik T, Kruger H. Toothy craniopharyngioma: a literature review and case report of craniopharyngioma with extensive odontogenic differentiation and tooth formation. Childs Nerv Syst. 2011;27:323–326. doi: 10.1007/s00381-010-1296-6.
    1. Muller HL, Merchant TE, Puget S, Martinez-Barbera JP. New outlook on the diagnosis, treatment and follow-up of childhood-onset craniopharyngioma. Nat Rev Endocrinol. 2017
    1. Neven B, Marvillet I, Terrada C, Ferster A, Boddaert N, Couloignier V, Pinto G, Pagnier A, Bodemer C, Bodaghi B, et al. Long-term efficacy of the interleukin-1 receptor antagonist anakinra in ten patients with neonatal-onset multisystem inflammatory disease/chronic infantile neurologic, cutaneous, articular syndrome. Arthritis Rheum. 2010;62:258–267. doi: 10.1002/art.25057.
    1. Paulus W, Stockel C, Krauss J, Sorensen N, Roggendorf W. Odontogenic classification of craniopharyngiomas: a clinicopathological study of 54 cases. Histopathology. 1997;30:172–176. doi: 10.1046/j.1365-2559.1997.d01-584.x.
    1. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747. doi: 10.1016/j.ccr.2014.04.021.
    1. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914.
    1. Robert C, Flaherty KT, Hersey P, Nathan PD, Garbe C, Milhem MM, Deminov LV, Hassel JC, Rutkowski P, Mohr P, et al. METRIC phase III study: efficacy of trametinib (T), a potent and selective MEK inhibitor (MEKi), in progression-free survival (PFS) and overall survival (OS), compared with chemotherapy (C) in patients (pts) with BRAFV600E/K mutant advanced or metastatic melanoma (MM) J Clin Oncol. 2012;30:LBA8509–LBA8509. doi: 10.1200/jco.2012.30.18_suppl.lba8509.
    1. Seemayer TA, Blundell JS, Wiglesworth FW. Pituitary craniopharyngioma with tooth formation. Cancer. 1972;29:423–430. doi: 10.1002/1097-0142(197202)29:2<423::AID-CNCR2820290225>;2-X.
    1. Sekine S, Sato S, Takata T, Fukuda Y, Ishida T, Kishino M, Shibata T, Kanai Y, Hirohashi S. Beta-catenin mutations are frequent in calcifying odontogenic cysts, but rare in ameloblastomas. Am J Pathol. 2003;163:1707–1712. doi: 10.1016/S0002-9440(10)63528-6.
    1. Sekine S, Takata T, Shibata T, Mori M, Morishita Y, Noguchi M, Uchida T, Kanai Y, Hirohashi S. Expression of enamel proteins and LEF1 in adamantinomatous craniopharyngioma: evidence for its odontogenic epithelial differentiation. Histopathology. 2004;45:573–579. doi: 10.1111/j.1365-2559.2004.02029.x.
    1. So A, De Smedt T, Revaz S, Tschopp J. A pilot study of IL-1 inhibition by anakinra in acute gout. Arthritis Res Ther. 2007;9:R28. doi: 10.1186/ar2143.
    1. Stache C, Holsken A, Schlaffer SM, Hess A, Metzler M, Frey B, Fahlbusch R, Flitsch J, Buchfelder M, Buslei R. Insights into the infiltrative behavior of adamantinomatous craniopharyngioma in a new xenotransplant mouse model. Brain Pathol. 2015;25:1–10. doi: 10.1111/bpa.12148.
    1. Tena-Suck ML, Salinas-Lara C, Arce-Arellano RI, Rembao-Bojorquez D, Morales-Espinosa D, Sotelo J, Arrieta O. Clinico-pathological and immunohistochemical characteristics associated to recurrence/regrowth of craniopharyngiomas. Clin Neurol Neurosurg. 2006;108:661–669. doi: 10.1016/j.clineuro.2006.01.007.
    1. Tucker A, Sharpe P. The cutting-edge of mammalian development; how the embryo makes teeth. Nat Rev Genet. 2004;5:499–508. doi: 10.1038/nrg1380.
    1. Tucker AS, Headon DJ, Schneider P, Ferguson BM, Overbeek P, Tschopp J, Sharpe PT. Edar/Eda interactions regulate enamel knot formation in tooth morphogenesis. Development. 2000;127:4691–4700.
    1. Wang Y, Li L, Zheng Y, Yuan G, Yang G, He F, Chen Y. BMP activity is required for tooth development from the lamina to bud stage. J Dent Res. 2012;91:690–695. doi: 10.1177/0022034512448660.
    1. Xavier GM, Patist AL, Healy C, Pagrut A, Carreno G, Sharpe PT, Martinez-Barbera JP, Thavaraj S, Cobourne MT, Andoniadou CL. Activated WNT signaling in postnatal SOX2-positive dental stem cells can drive odontoma formation. Sci Rep. 2015;5:14479. doi: 10.1038/srep14479.
    1. Zhou J, Zhang C, Pan J, Chen L, Qi ST. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human adamantinomatous craniopharyngioma cells and promotes tumor cell migration. Mol Med Rep. 2017;15:4123–4131. doi: 10.3892/mmr.2017.6538.

Source: PubMed

3
S'abonner