Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Cell Commun Signal ; 16(4): 637-648, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35102500

ABSTRACT

Recent advancements in 3D in vitro culture have allowed for the development of cancer tissue models which accurately recapitulate the tumour microenvironment. Consequently, there has been increased innovation in therapeutic drug screening. While organoid cultures show great potential, they are limited by the time scale of their growth in vitro and the dependence upon commercial matrices, such as Matrigel, which do not allow for manipulations of their composition or mechanical properties. Here, we show a straightforward approach for the isolation and culture of primary human renal carcinoma cells and matched non-affected kidney. This approach does not require any specific selection for cancer cells, and allows for their direct culture in amenable 3D collagen-based matrices, with the preservation of cancer cells as confirmed by NGS sequencing. This method allows for culture of patient-derived cancer cells in 3D microenvironment, which can be used for downstream experimentation such as investigation of cell-matrix interaction or drug screening.

2.
Br J Cancer ; 117(3): 398-408, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28632727

ABSTRACT

BACKGROUND: The most prevalent form of bone cancer is osteosarcoma (OS), which is associated with poor prognosis in case of metastases formation. Mice harbouring liver kinase B1 (LKB1+/-) develop osteoblastoma-like tumours. Therefore, we asked whether loss of LKB1 gene has a role in the pathogenesis of human OS. METHODS: Osteosarcomas (n=259) were screened for LKB1 and sirtuin 1 (SIRT1) protein expression using immunohistochemistry and western blot. Those cases were also screened for LKB1 genetic alterations by next-generation sequencing, Sanger sequencing, restriction fragment length polymorphism and fluorescence in situ hybridisation approaches. We studied LKB1 protein degradation through SIRT1 expression. MicroRNA expression investigations were also conducted to identify the microRNAs involved in the SIRT1/LKB1 pathway. RESULTS: Forty-one per cent (106 out of 259) OS had lost LKB1 protein expression with no evident genetic anomalies. We obtained evidence that SIRT1 impairs LKB1 protein stability, and that SIRT1 depletion leads to accumulation of LKB1 in OS cell lines resulting in growth arrest. Further investigations revealed the role of miR-204 in the regulation of SIRT1 expression, which impairs LKB1 stability. CONCLUSIONS: We demonstrated the involvement of sequential regulation of miR-204/SIRT1/LKB1 in OS cases and showed a mechanism for the loss of expression of LKB1 tumour suppressor in this malignancy.


Subject(s)
Bone Neoplasms/genetics , Osteosarcoma/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Sirtuin 1/metabolism , AMP-Activated Protein Kinase Kinases , Anoikis/genetics , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Bone Neoplasms/metabolism , Bortezomib/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Humans , MicroRNAs/genetics , Naphthols/pharmacology , Osteosarcoma/metabolism , Protein Biosynthesis/drug effects , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , TOR Serine-Threonine Kinases/metabolism , Transfection
3.
Virchows Arch ; 467(2): 203-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25838078

ABSTRACT

Desmoid-type fibromatoses are locally aggressive and frequently recurrent tumours, and an accurate diagnosis is essential for patient management. The majority of sporadic lesions harbour beta-catenin (CTNNB1) mutations. We used next-generation sequencing to detect CTNNB1 mutations and to compare the sensitivity and specificity of next-generation sequencing with currently employed mutation detection techniques: mutation-specific restriction enzyme digestion and polymerase chain reaction amplification. DNA was extracted from formalin-fixed paraffin-embedded needle biopsy or resection tissue sections from 144 patients with sporadic desmoid-type fibromatoses, four patients with syndrome-related desmoid-type fibromatoses and 11 morphological mimics. Two primer pairs were designed for CTNNB1 mutation hotspots. Using ≥10 ng of DNA, libraries were generated by Fluidigm and sequenced on the Ion Torrent Personal Genome Machine. Next-generation sequencing had a sensitivity of 92.36 % (133/144, 95 % CIs: 86.74 to 96.12 %) and a specificity of 100 % for the detection of CTNNB1 mutations in desmoid-type fibromatoses-like spindle cell lesions. All mutations detected by mutation-specific restriction enzyme digestion were identified by next-generation sequencing. Next-generation sequencing identified additional mutations in 11 tumours that were not detected by mutation-specific restriction enzyme digestion, two of which have not been previously described. Next-generation sequencing is highly sensitive for the detection of CTNNB1 mutations. This multiplex assay has the advantage of detecting additional mutations compared to those detected by mutation-specific restriction enzyme digestion (sensitivity 82.41 %). The technology requires minimal DNA and is time- and cost-efficient.


Subject(s)
DNA Mutational Analysis/methods , Fibromatosis, Aggressive/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , beta Catenin/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Young Adult
4.
J Pathol Clin Res ; 1(2): 113-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-27499898

ABSTRACT

Driver mutations in the two histone 3.3 (H3.3) genes, H3F3A and H3F3B, were recently identified by whole genome sequencing in 95% of chondroblastoma (CB) and by targeted gene sequencing in 92% of giant cell tumour of bone (GCT). Given the high prevalence of these driver mutations, it may be possible to utilise these alterations as diagnostic adjuncts in clinical practice. Here, we explored the spectrum of H3.3 mutations in a wide range and large number of bone tumours (n = 412) to determine if these alterations could be used to distinguish GCT from other osteoclast-rich tumours such as aneurysmal bone cyst, nonossifying fibroma, giant cell granuloma, and osteoclast-rich malignant bone tumours and others. In addition, we explored the driver landscape of GCT through whole genome, exome and targeted sequencing (14 gene panel). We found that H3.3 mutations, namely mutations of glycine 34 in H3F3A, occur in 96% of GCT. We did not find additional driver mutations in GCT, including mutations in IDH1, IDH2, USP6, TP53. The genomes of GCT exhibited few somatic mutations, akin to the picture seen in CB. Overall our observations suggest that the presence of H3F3A p.Gly34 mutations does not entirely exclude malignancy in osteoclast-rich tumours. However, H3F3A p.Gly34 mutations appear to be an almost essential feature of GCT that will aid pathological evaluation of bone tumours, especially when confronted with small needle core biopsies. In the absence of H3F3A p.Gly34 mutations, a diagnosis of GCT should be made with caution.

7.
Cancer Med ; 3(4): 980-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24861215

ABSTRACT

Osteosarcoma, the most common primary bone sarcoma, is a genetically complex disease with no widely accepted biomarker to allow stratification of patients for treatment. After a recent report of one osteosarcoma cell line and one tumor exhibiting fibroblastic growth factor receptor 1 (FGFR1) gene amplification, the aim of this work was to assess the frequency of FGFR1 amplification in a larger cohort of osteosarcoma and to determine if this biomarker could be used for stratification of patients for treatment. About 352 osteosarcoma samples from 288 patients were analyzed for FGFR1 amplification by interphase fluorescence in situ hybridization. FGFR1 amplification was detected in 18.5% of patients whose tumors revealed a poor response to chemotherapy, and no patients whose tumors responded well to therapy harbored this genetic alteration. FGFR1 amplification is present disproportionately in the rarer histological variants of osteosarcoma. This study provides a rationale for inclusion of patients with osteosarcoma in clinical trials using FGFR kinase inhibitors.


Subject(s)
Bone Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Osteosarcoma/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adolescent , Adult , Aged , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Chemotherapy, Adjuvant , Female , Gene Amplification , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoadjuvant Therapy , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Retrospective Studies , Treatment Outcome , Young Adult
8.
Genome Med ; 6(12): 116, 2014.
Article in English | MEDLINE | ID: mdl-25587359

ABSTRACT

BACKGROUND: The use of tumour xenografts is a well-established research tool in cancer genomics but has not yet been comprehensively evaluated for cancer epigenomics. METHODS: In this study, we assessed the suitability of patient-derived tumour xenografts (PDXs) for methylome analysis using Infinium 450 K Beadchips and MeDIP-seq. RESULTS: Controlled for confounding host (mouse) sequences, comparison of primary PDXs and matching patient tumours in a rare (osteosarcoma) and common (colon) cancer revealed that an average 2.7% of the assayed CpG sites undergo major (Δß ≥ 0.51) methylation changes in a cancer-specific manner as a result of the xenografting procedure. No significant subsequent methylation changes were observed after a second round of xenografting between primary and secondary PDXs. Based on computational simulation using publically available methylation data, we additionally show that future studies comparing two groups of PDXs should use 15 or more samples in each group to minimise the impact of xenografting-associated changes in methylation on comparison results. CONCLUSIONS: Our results from rare and common cancers indicate that PDXs are a suitable discovery tool for cancer epigenomics and we provide guidance on how to overcome the observed limitations.

9.
Nat Genet ; 45(12): 1479-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24162739

ABSTRACT

It is recognized that some mutated cancer genes contribute to the development of many cancer types, whereas others are cancer type specific. For genes that are mutated in multiple cancer classes, mutations are usually similar in the different affected cancer types. Here, however, we report exquisite tumor type specificity for different histone H3.3 driver alterations. In 73 of 77 cases of chondroblastoma (95%), we found p.Lys36Met alterations predominantly encoded in H3F3B, which is one of two genes for histone H3.3. In contrast, in 92% (49/53) of giant cell tumors of bone, we found histone H3.3 alterations exclusively in H3F3A, leading to p.Gly34Trp or, in one case, p.Gly34Leu alterations. The mutations were restricted to the stromal cell population and were not detected in osteoclasts or their precursors. In the context of previously reported H3F3A mutations encoding p.Lys27Met and p.Gly34Arg or p.Gly34Val alterations in childhood brain tumors, a remarkable picture of tumor type specificity for histone H3.3 driver alterations emerges, indicating that histone H3.3 residues, mutations and genes have distinct functions.


Subject(s)
Bone Neoplasms/genetics , Chondroblastoma/genetics , Giant Cell Tumor of Bone/genetics , Histones/genetics , Amino Acid Sequence , Bone Neoplasms/epidemiology , Case-Control Studies , Cells, Cultured , Child , Chondroblastoma/epidemiology , Gene Frequency , Giant Cell Tumor of Bone/epidemiology , Humans , Mutation
11.
Nat Genet ; 44(11): 1185-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064415

ABSTRACT

Chordoma is a rare malignant bone tumor that expresses the transcription factor T. We conducted an association study of 40 individuals with chordoma and 358 ancestry-matched controls, with replication in an independent cohort. Whole-exome and Sanger sequencing of T exons showed strong association of the common nonsynonymous SNP rs2305089 with chordoma risk (allelic odds ratio (OR) = 6.1, 95% confidence interval (CI) = 3.1-12.1; P = 4.4 × 10(-9)), a finding that is exceptional in cancers with a non-Mendelian mode of inheritance.


Subject(s)
Bone Neoplasms/genetics , Chordoma/genetics , Fetal Proteins/genetics , T-Box Domain Proteins/genetics , Exome , Humans , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , White People/genetics
12.
J Pathol ; 228(3): 274-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22847733

ABSTRACT

Chordoma is a rare malignant tumour of bone, the molecular marker of which is the expression of the transcription factor, brachyury. Having recently demonstrated that silencing brachyury induces growth arrest in a chordoma cell line, we now seek to identify its downstream target genes. Here we use an integrated functional genomics approach involving shRNA-mediated brachyury knockdown, gene expression microarray, ChIP-seq experiments, and bioinformatics analysis to achieve this goal. We confirm that the T-box binding motif of human brachyury is identical to that found in mouse, Xenopus, and zebrafish development, and that brachyury acts primarily as an activator of transcription. Using human chordoma samples for validation purposes, we show that brachyury binds 99 direct targets and indirectly influences the expression of 64 other genes, thereby acting as a master regulator of an elaborate oncogenic transcriptional network encompassing diverse signalling pathways including components of the cell cycle, and extracellular matrix components. Given the wide repertoire of its active binding and the relative specific localization of brachyury to the tumour cells, we propose that an RNA interference-based gene therapy approach is a plausible therapeutic avenue worthy of investigation.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/physiopathology , Chordoma/genetics , Chordoma/physiopathology , Fetal Proteins/genetics , Fetal Proteins/physiology , Genomics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Animals , Bone Neoplasms/pathology , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation , Chordoma/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Genetic Therapy , Humans , Mice , Notochord/pathology , RNA Interference , Transcription, Genetic/physiology , Xenopus , Zebrafish
14.
Nat Genet ; 43(12): 1262-5, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057236

ABSTRACT

Ollier disease and Maffucci syndrome are characterized by multiple central cartilaginous tumors that are accompanied by soft tissue hemangiomas in Maffucci syndrome. We show that in 37 of 40 individuals with these syndromes, at least one tumor has a mutation in isocitrate dehydrogenase 1 (IDH1) or in IDH2, 65% of which result in a R132C substitution in the protein. In 18 of 19 individuals with more than one tumor analyzed, all tumors from a given individual shared the same IDH1 mutation affecting Arg132. In 2 of 12 subjects, a low level of mutated DNA was identified in non-neoplastic tissue. The levels of the metabolite 2HG were measured in a series of central cartilaginous and vascular tumors, including samples from syndromic and nonsyndromic subjects, and these levels correlated strongly with the presence of IDH1 mutations. The findings are compatible with a model in which IDH1 or IDH2 mutations represent early post-zygotic occurrences in individuals with these syndromes.


Subject(s)
Enchondromatosis/genetics , Isocitrate Dehydrogenase/genetics , Mutation, Missense , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Genetic Association Studies , Humans , Male , Middle Aged , Mosaicism , Sequence Analysis, DNA , Young Adult
15.
J Pathol ; 224(3): 334-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21598255

ABSTRACT

Somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 occur in gliomas and acute myeloid leukaemia (AML). Since patients with multiple enchondromas have occasionally been reported to have these conditions, we hypothesized that the same mutations would occur in cartilaginous neoplasms. Approximately 1200 mesenchymal tumours, including 220 cartilaginous tumours, 222 osteosarcomas and another ∼750 bone and soft tissue tumours, were screened for IDH1 R132 mutations, using Sequenom(®) mass spectrometry. Cartilaginous tumours and chondroblastic osteosarcomas, wild-type for IDH1 R132, were analysed for IDH2 (R172, R140) mutations. Validation was performed by capillary sequencing and restriction enzyme digestion. Heterozygous somatic IDH1/IDH2 mutations, which result in the production of a potential oncometabolite, 2-hydroxyglutarate, were only detected in central and periosteal cartilaginous tumours, and were found in at least 56% of these, ∼40% of which were represented by R132C. IDH1 R132H mutations were confirmed by immunoreactivity for this mutant allele. The ratio of IDH1:IDH2 mutation was 10.6 : 1. No IDH2 R140 mutations were detected. Mutations were detected in enchondromas through to conventional central and dedifferentiated chondrosarcomas, in patients with both solitary and multiple neoplasms. No germline mutations were detected. No mutations were detected in peripheral chondrosarcomas and osteochondromas. In conclusion, IDH1 and IDH2 mutations represent the first common genetic abnormalities to be identified in conventional central and periosteal cartilaginous tumours. As in gliomas and AML, the mutations appear to occur early in tumourigenesis. We speculate that a mosaic pattern of IDH-mutation-bearing cells explains the reports of diverse tumours (gliomas, AML, multiple cartilaginous neoplasms, haemangiomas) occurring in the same patient.


Subject(s)
Bone Neoplasms/genetics , Chondroma/genetics , Chondrosarcoma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Bone Neoplasms/diagnosis , Bone Neoplasms/pathology , Chondroma/diagnosis , Chondroma/pathology , Chondrosarcoma/pathology , Enchondromatosis/genetics , Enchondromatosis/pathology , Female , Follow-Up Studies , Germ-Line Mutation , Humans , Magnetic Resonance Imaging , Male , Osteosarcoma/genetics , Osteosarcoma/pathology
16.
Genome Res ; 21(4): 515-24, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21324880

ABSTRACT

Aberrant DNA methylation (DNAm) was first linked to cancer over 25 yr ago. Since then, many studies have associated hypermethylation of tumor suppressor genes and hypomethylation of oncogenes to the tumorigenic process. However, most of these studies have been limited to the analysis of promoters and CpG islands (CGIs). Recently, new technologies for whole-genome DNAm (methylome) analysis have been developed, enabling unbiased analysis of cancer methylomes. By using MeDIP-seq, we report a sequencing-based comparative methylome analysis of malignant peripheral nerve sheath tumors (MPNSTs), benign neurofibromas, and normal Schwann cells. Analysis of these methylomes revealed a complex landscape of DNAm alterations. In contrast to what has been reported for other tumor types, no significant global hypomethylation was observed in MPNSTs using methylome analysis by MeDIP-seq. However, a highly significant (P < 10(-100)) directional difference in DNAm was found in satellite repeats, suggesting these repeats to be the main target for hypomethylation in MPNSTs. Comparative analysis of the MPNST and Schwann cell methylomes identified 101,466 cancer-associated differentially methylated regions (cDMRs). Analysis showed these cDMRs to be significantly enriched for two satellite repeat types (SATR1 and ARLα) and suggests an association between aberrant DNAm of these sequences and transition from healthy cells to malignant disease. Significant enrichment of hypermethylated cDMRs in CGI shores (P < 10(-60)), non-CGI-associated promoters (P < 10(-4)) and hypomethylated cDMRs in SINE repeats (P < 10(-100)) was also identified. Integration of DNAm and gene expression data showed that the expression pattern of genes associated with CGI shore cDMRs was able to discriminate between disease phenotypes. This study establishes MeDIP-seq as an effective method to analyze cancer methylomes.


Subject(s)
DNA Methylation/genetics , Epigenomics , Nerve Sheath Neoplasms/genetics , Cluster Analysis , CpG Islands/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Minisatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis/methods
17.
J Pathol ; 223(3): 327-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21171078

ABSTRACT

A variety of analyses, including fluorescence in situ hybridization (FISH), quantitative PCR (qPCR) and array CGH (aCGH), have been performed on a series of chordomas from 181 patients. Twelve of 181 (7%) tumours displayed amplification of the T locus and an additional two cases showed focal amplification; 70/181 (39%) tumours were polysomic for chromosome 6, and 8/181 (4.5%) primary tumours showed a minor allelic gain of T as assessed by FISH. No germline alteration of the T locus was identified in non-neoplastic tissue from 40 patients. Copy number gain of T was seen in a similar percentage of sacrococcygeal, mobile spine and base of skull tumours. Knockdown of T in the cell line, U-CH1, which showed polysomy of chromosome 6 involving 6q27, resulted in a marked decrease in cell proliferation and morphological features consistent with a senescence-like phenotype. The U-CH1 cell line was validated as representing chordoma by the generation of xenografts, which showed typical chordoma morphology and immunohistochemistry in the NOD/SCID/interleukin 2 receptor [IL2r]gammanull mouse model. In conclusion, chromosomal aberrations resulting in gain of the T locus are common in sporadic chordomas and expression of this gene is critical for proliferation of chordoma cells in vitro.


Subject(s)
Chordoma/genetics , Fetal Proteins/genetics , T-Box Domain Proteins/genetics , Animals , Cell Proliferation , Chordoma/metabolism , Chordoma/pathology , Chromosome Aberrations , DNA Copy Number Variations , DNA, Neoplasm/genetics , Fetal Proteins/metabolism , Gene Knockdown Techniques , Genetic Predisposition to Disease , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Polymerase Chain Reaction/methods , T-Box Domain Proteins/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
18.
J Pathol ; 223(3): 336-46, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21171079

ABSTRACT

Chordoma, the molecular hallmark of which is T (brachyury), is a rare malignant bone tumour with a high risk of local recurrence and a tumour from which metastatic disease is a common late event. Currently, there is no effective drug therapy for treating chordomas, although there is evidence that some patients respond to the empirical use of epidermal growth factor receptor (EGFR) antagonists. The aim of this study was to determine the role of EGFR in the pathogenesis of chordoma. Paraffin-embedded material from 173 chordomas from 160 patients [sacro-coccygeal (n = 94), skull-based (n = 50), and mobile spine (n = 16)] was analysed by immunohistochemistry and revealed total EGFR expression in 69% of cases analysed. Of 147 informative chordomas analysed by FISH, 38% revealed high-level EGFR polysomy, 4% high-level polysomy with focal amplification, 18% low-level polysomy, and 39% disomy. Phospho-receptor tyrosine kinase array membranes showed EGFR activation in the chordoma cell line U-CH1 and all of the three chordomas analysed. Direct sequencing of EGFR (exons 18-21), KRAS, NRAS, HRAS (exons 2, 3), and BRAF (exons 11, 15) using DNA from 62 chordomas failed to reveal mutations. PTEN expression was absent by immunohistochemistry in 19 of 147 (13%) analysed chordomas, only one of which revealed high-level polysomy of EGFR. The EGFR inhibitor tyrphostin (AG 1478) markedly inhibited proliferation of the chordoma cell line U-CH1 in vitro and diminished EGFR phosphorylation in a dose-dependant manner, a finding supported by inhibition of phosphorylated Erk1/2. p-Akt was suppressed to a much lesser degree in these experiments. There was no reduction of T as assessed by western blotting. These data implicate aberrant EGFR signalling in the pathogenesis of chordoma. This study provides a strategy for patient stratification for treatment with EGFR antagonists.


Subject(s)
Bone Neoplasms/metabolism , Chordoma/metabolism , ErbB Receptors/metabolism , Antineoplastic Agents/pharmacology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Chordoma/genetics , Chordoma/pathology , DNA Mutational Analysis/methods , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , In Situ Hybridization, Fluorescence , Mutation , Neoplasm Proteins/metabolism , Quinazolines , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Skull Base Neoplasms/metabolism , Tumor Cells, Cultured , Tyrphostins/pharmacology
19.
Skeletal Radiol ; 39(1): 63-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19830424

ABSTRACT

OBJECTIVE: To report on the biochemistry and clinical and genetic findings of two siblings, the younger sister presenting with recurrent bone pain of the radius and ulna, and medullary sclerosis, and the older brother with soft tissue calcific deposits (tumoral calcinosis) but who later developed bone pain. Both were found to be hyperphosphaturic. MATERIALS AND METHODS: The index family comprised four individuals (father, mother, brother, sister). The affected siblings were the offspring of a non-consanguineous Indian family of Tamil origin. Bidirectional sequencing was performed on the DNA from the index family and on 160 alleles from a population of 80 unrelated unaffected control individuals of Tamil extraction and 72 alleles from individuals of non-Tamil origin. RESULTS: Two symptomatic siblings were found to harbour previously unreported compound heterozygous missense UDP-N-acetyl-D-galactosamine: polypeptide N-acetylgalactosaminyltransferase 3 (GalNAc-transferase; GALNT3) mutations in exon 4 c.842A>G and exon 5 c.1097T>G. This sequence variation was not detected in the control DNA. This is the first report of siblings exhibiting stigmata of familial tumoral calcinosis and hyperostosis-hyperphosphataemia syndrome with documented evidence of autosomal recessive missense GALNT3 mutations. CONCLUSION: The findings from this family add further evidence to the literature that familial tumoral calcinosis and hyperostosis-hyperphosphataemia syndrome are manifestations of the same disease and highlight the importance of appropriate metabolic and genetic investigations.


Subject(s)
Bone Neoplasms/genetics , Calcinosis/genetics , Hyperphosphatemia/genetics , N-Acetylgalactosaminyltransferases/genetics , Adolescent , Bone Neoplasms/diagnostic imaging , Calcinosis/diagnostic imaging , Female , Genetic Predisposition to Disease , Humans , Hyperphosphatemia/diagnostic imaging , Male , Mutation, Missense , Polymerase Chain Reaction , Radiography , Polypeptide N-acetylgalactosaminyltransferase
20.
Mod Pathol ; 22(8): 996-1005, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19407855

ABSTRACT

Chordomas are rare primary malignant bone tumours that derive from notochord precursor cells and express brachyury, a molecule involved in notochord development. Little is known about the genetic events responsible for driving the growth of this tumour, but it is well established that brachyury is regulated through fibroblastic growth factor receptors (FGFRs) through RAS/RAF/MEK/ERK and ETS2 in ascidian, Xenopus and zebrafish, although little is known about its regulation in mammals. The aim of this study was to attempt to identify the molecular genetic events that are responsible for the pathogenesis of chordomas with particular focus on the FGFR signalling pathway on the basis of the evidence in the ascidian and Xenopus models that the expression of brachyury requires the activation of this pathway. Immunohistochemistry showed that 47 of 50 chordomas (94%) expressed at least one of the FGFRs, and western blotting showed phosphorylation of fibroblast growth factor receptor substrate 2 alpha (FRS2alpha), an adaptor signalling protein, that links FGFR to the RAS/RAF/MEK/ERK pathway. Screening for mutations in brachyury (all coding exons and promoter), FGFRs 1-4 (previously reported mutations), KRAS (codons 12, 13, 51, 61) and BRAF (exons 11 and 15) failed to show any genetic alterations in 23 chordomas. Fluorescent in situ hybridisation analysis on FGFR4, ETS2 and brachyury failed to show either amplification of these genes, although there was minor allelic gain in brachyury in three tumours, or translocation for ERG and ETS2 loci. The key genetic events responsible for the initiation and progression of chordomas remain to be discovered.


Subject(s)
Chordoma/genetics , Fetal Proteins/genetics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction/physiology , Spinal Neoplasms/genetics , T-Box Domain Proteins/genetics , Adult , Aged , Aged, 80 and over , Blotting, Western , Chordoma/metabolism , Chromatography, High Pressure Liquid , DNA Mutational Analysis , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fetal Proteins/metabolism , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Middle Aged , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Spinal Neoplasms/metabolism , T-Box Domain Proteins/metabolism , Tissue Array Analysis , Young Adult , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...