Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Genes Chromosomes Cancer ; 63(5): e23241, 2024 05.
Article in English | MEDLINE | ID: mdl-38738966

ABSTRACT

Osteoblastomas (OBs) are benign neoplasms constituting approximately 1% of primary bone tumors with a predilection for the spine and sacrum. We describe an OB of the proximal phalanx of the left thumb in a 38-year-old female. MRI of left hand demonstrated a 29-mm mildly expansile enhancing lesion involving the entire proximal phalanx of the first digit. Histology displayed a bone-forming tumor consisting of trabeculae of remodeled woven bone framed by plump osteoblasts in a vascularized background. Next-generation sequencing analysis identified a PRSS44::ALK fusion gene.


Subject(s)
Bone Neoplasms , Osteoblastoma , Thumb , Humans , Female , Adult , Thumb/pathology , Thumb/abnormalities , Osteoblastoma/genetics , Osteoblastoma/pathology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Oncogene Proteins, Fusion/genetics
2.
J Clin Pathol ; 76(11): 721-726, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37553246

ABSTRACT

FOS and FOSB proto-oncogens are involved in a wide variety of tumourigenic processes. FOS and FOSB gene rearrangements are observed in epithelioid haemangioma, pseudomyogenic haemangioendothelioma, osteoid osteoma/osteoblastoma/cementoblastoma and proliferative myositis/fasciitis. In this review, we provide an overview of FOS and FOSB, including their functions and the differences between lesions with known FOS/FOSB gene rearrangements. Additionally, we discuss the use of FOS/FOSB immunohistochemistry as a diagnostic tool for these lesions.


Subject(s)
Proto-Oncogene Proteins c-fos , Humans , Bone Neoplasms/pathology , Cell Transformation, Neoplastic , Osteoblastoma/diagnosis , Osteoblastoma/genetics , Osteoblastoma/pathology , Proto-Oncogene Proteins c-fos/genetics , Soft Tissue Neoplasms/pathology
3.
Mod Pathol ; 35(9): 1204-1211, 2022 09.
Article in English | MEDLINE | ID: mdl-35347251

ABSTRACT

Rearrangements of the transcription factors FOS and FOSB have recently been identified as the genetic driver event underlying osteoid osteoma and osteoblastoma. Nuclear overexpression of FOS and FOSB have since then emerged as a reliable surrogate marker despite limitations in specificity and sensitivity. Indeed, osteosarcoma can infrequently show nuclear FOS expression and a small fraction of osteoblastomas seem to arise independent of FOS/FOSB rearrangements. Acid decalcification and tissue preservation are additional factors that can negatively influence immunohistochemical testing and make diagnostic decision-making challenging in individual cases. Particularly aggressive appearing osteoblastomas, also referred to as epithelioid osteoblastomas, and osteoblastoma-like osteosarcoma can be difficult to distinguish, underlining the need for additional markers to support the diagnosis. Methylation and copy number profiling, a technique well established for the classification of brain tumors, might fill this gap. Here, we set out to comprehensively characterize a series of 77 osteoblastomas by immunohistochemistry, fluorescence in-situ hybridization as well as copy number and methylation profiling and compared our findings to histologic mimics. Our results show that osteoblastomas are uniformly characterized by flat copy number profiles that can add certainty in reaching the correct diagnosis. The methylation cluster formed by osteoblastomas, however, so far lacks specificity and can be misleading in individual cases.


Subject(s)
Bone Neoplasms , Osteoblastoma , Osteosarcoma , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , DNA Copy Number Variations , Humans , Methylation , Osteoblastoma/diagnosis , Osteoblastoma/genetics , Osteoblastoma/metabolism , Osteosarcoma/pathology
4.
Skeletal Radiol ; 51(3): 673-679, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34448010

ABSTRACT

Epithelioid osteoblastoma, sometimes equated with aggressive osteoblastoma, is a variant of osteoblastoma that typically demonstrates more worrisome imaging and pathological features compared to conventional osteoblastoma. These more aggressive features can overlap with those seen in osteosarcoma, creating a diagnostic challenge for radiologists and pathologists. Recent identification of FOS and FOSB gene rearrangements in osteoid osteoma and osteoblastoma has allowed for greater diagnostic confidence following biopsy, but careful radiological-pathological correlation remains a key component for guiding appropriate management. Although the imaging features of conventional osteoblastoma have been previously described, there are limited examples in the literature of the imaging appearance of epithelioid osteoblastoma, and none with secondary aneurysmal bone cyst. In this case report, we detail the clinical, imaging, and histological characteristics of a proximal femoral epithelioid osteoblastoma which was pathologically confirmed by FOS and FOSB genetic testing. The initial imaging impression favored a malignancy, but when the biopsy results were correlated in a multidisciplinary fashion with the imaging, epithelioid osteoblastoma became the leading diagnosis which was subsequently genetically confirmed. This case emphasizes the value of multidisciplinary radiology-pathology correlation in routine practice.


Subject(s)
Bone Cysts, Aneurysmal , Bone Neoplasms , Osteoblastoma , Osteoma, Osteoid , Bone Cysts, Aneurysmal/diagnostic imaging , Bone Cysts, Aneurysmal/genetics , Bone Cysts, Aneurysmal/surgery , Bone Neoplasms/complications , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/genetics , Gene Rearrangement , Humans , Osteoblastoma/diagnostic imaging , Osteoblastoma/genetics , Osteoblastoma/surgery , Osteoma, Osteoid/complications , Osteoma, Osteoid/diagnostic imaging , Osteoma, Osteoid/genetics
5.
Life Sci ; 288: 118993, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-33545202

ABSTRACT

OBJECTIVE: To study the proapoptotic effect of ligustilide on osteoblastoma (OS) and the relative related molecular mechanism. METHODS AND MATERIALS: An MTT was used to examine the proliferation of OS cells, and Flow cytometry was used to analyze apoptosis and the cell cycle. Western blotting was used to detect the signaling pathway of apoptosis, and immunohistochemical (IH) staining was used to detect the apoptosis status of OS cells. A TLR4 inhibitor was used to study the effect of ligustilide on OS. RESULTS: Ligustilide inhibited OS cell proliferation but had no inhibitory effect on normal bone marrow cells. Flow cytometry results showed that ligustilide induced apoptosis in OS cells, and the cell cycle was arrested at the M/G2 phase. Western blot results showed that ERK, P53, P21, Caspase 9, Caspase 8 and Caspase 3 were all activated; cytochrome C and Bax increased; and Bcl-2 decreased when OS was treated with ligustilide. When an ERK or Caspase inhibitor was added to the culture medium, the apoptosis of OS cells decreased to some degree. When OS cells were pretreated with CLI-095, which is a TLR4 inhibitor, the percentage of apoptotic cells and cell cycle arrest were both reversed. IH results also showed that ligustilide induced apoptosis in OS cells, and the effect was blocked by the TLR4 inhibitor. CONCLUSION: Ligustilide selectively inhibited the proliferation of OS cells by inducing apoptosis, which possibly included endogenous and exogenous apoptosis through TLR4.


Subject(s)
4-Butyrolactone/analogs & derivatives , Bone Neoplasms/drug therapy , Caspases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Signaling System/drug effects , Osteoblastoma/drug therapy , Toll-Like Receptor 4/metabolism , 4-Butyrolactone/pharmacology , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Caspases/genetics , Cell Cycle Checkpoints , Cell Movement , Cell Proliferation , Humans , Osteoblastoma/genetics , Osteoblastoma/metabolism , Osteoblastoma/pathology , Toll-Like Receptor 4/genetics , Tumor Cells, Cultured
6.
Am J Surg Pathol ; 45(5): 690-693, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33653978

ABSTRACT

Cementoblastomas are rare odontogenic tumors developing in close proximity to the roots of teeth. Due to their striking morphologic resemblance to osteoblastomas of the peripheral skeleton, we set out to determine whether cementoblastomas harbor the same FOS rearrangements with overexpression of c-FOS as has recently been described for osteoblastomas. In total, 16 cementoblastomas were analyzed for FOS expression by immunohistochemistry and for FOS rearrangements by fluorescence in situ hybridization (FISH). We observed strong and diffuse staining of c-FOS in 71% of cementoblastomas and identified a FOS rearrangement in all cases (n=3) applicable for FISH. In the remaining cases, FISH failed due to decalcification. Cementoblastomas harbor similar FOS rearrangements and show overexpression of c-FOS like osteoblastomas, suggesting that both entities might represent parts of the spectrum of the same disease.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Dental Cementum , Gene Rearrangement , Odontogenic Tumors , Osteoblastoma , Proto-Oncogene Proteins c-fos , Tooth Root , Adolescent , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Bone Neoplasms/chemistry , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Child , Dental Cementum/chemistry , Dental Cementum/pathology , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Netherlands , Odontogenic Tumors/chemistry , Odontogenic Tumors/genetics , Odontogenic Tumors/pathology , Osteoblastoma/chemistry , Osteoblastoma/genetics , Osteoblastoma/pathology , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/genetics , Switzerland , Tooth Root/chemistry , Tooth Root/pathology , Young Adult
7.
J Pathol Clin Res ; 6(4): 231-237, 2020 10.
Article in English | MEDLINE | ID: mdl-32542935

ABSTRACT

Osteoblastoma is a locally aggressive tumour of bone. Until recently, its underlying genetic features were largely unknown. During the past two years, reports have demonstrated that acquired structural variations affect the transcription factor FOS in a high proportion of cases. These rearrangements modify the terminal exon of the gene and are believed to stabilise both the FOS transcript and the encoded protein, resulting in high expression levels. Here, we applied in-depth genetic analyses to a series of 29 osteoblastomas, including five classified as epithelioid osteoblastoma. We found recurrent homozygous deletions of the NF2 gene in three of the five epithelioid cases and in one conventional osteoblastoma. These events were mutually exclusive from FOS mutations. Structural variations were determined by deep whole genome sequencing and the number of FOS-rearranged cases was less than previously reported (10/23, 43%). One conventional osteoblastoma displayed a novel mechanism of FOS upregulation; bringing the entire FOS gene under the control of the WNT5A enhancer that is itself activated by FOS. Taken together, we show that NF2 loss characterises a subgroup of osteoblastomas, distinct from FOS-rearranged cases. Both NF2 and FOS are involved in regulating bone homeostasis, thereby providing a mechanistic link to the excessive bone growth of osteoblastoma.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Gene Deletion , Gene Rearrangement , Neurofibromin 2/genetics , Osteoblastoma/genetics , Proto-Oncogene Proteins c-fos/genetics , Adolescent , Adult , Bone Neoplasms/pathology , Child , Child, Preschool , Enhancer Elements, Genetic , Epithelioid Cells/pathology , Europe , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Osteoblastoma/pathology , Osteogenesis , Phenotype , Wnt-5a Protein/genetics , Young Adult
8.
Cancer Genomics Proteomics ; 17(2): 161-168, 2020.
Article in English | MEDLINE | ID: mdl-32108038

ABSTRACT

BACKGROUND/AIM: Osteoblastoma is a rare benign tumor of the bones in which recurrent rearrangements of FOS have been found. Our aim was to investigate two osteoblastomas for possible genetic aberrations. MATERIALS AND METHODS: Cytogenetic, RNA sequencing, and molecular analyses were performed. RESULTS: A FOS-ANKH transcript was found in the first tumor, whereas a FOS-RUNX2 was detected in the second. Exon 4 of FOS fused with sequences either from intron 1 of ANKH or intron 5 of RUNX2. The fusion events introduced a stop codon and removed sequences involved in the regulation of FOS. CONCLUSION: Rearrangements and fusions of FOS show similarities with those of HMGA2 (a feature of leiomyomas and lipomas) and CSF1 (tenosynovial giant cell tumors). The replacement of a 3'-untranslated region, controlling the gene's expression, by a new sequence is thus a common pathogenetic theme shared by FOS, HMGA2, and CSF1 in many benign connective tissue tumors.


Subject(s)
Bone Neoplasms/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Osteoblastoma/genetics , Phosphate Transport Proteins/genetics , Base Sequence , Bone Neoplasms/metabolism , Child , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Gene Expression , Humans , Karyotype , Male , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Osteoblastoma/metabolism , Phosphate Transport Proteins/metabolism
9.
Virchows Arch ; 476(1): 147-157, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31741049

ABSTRACT

Bone tumours are difficult to diagnose and treat, as they are rare and over 60 different subtypes are recognised. The emergence of next-generation sequencing has partly elucidated the molecular mechanisms behind these tumours, including the group of bone forming tumours (osteoma, osteoid osteoma, osteoblastoma and osteosarcoma). Increased knowledge on the molecular mechanism could help to identify novel diagnostic markers and/or treatment options. Osteoid osteoma and osteoblastoma are bone forming tumours without malignant potential that have overlapping morphology. They were recently shown to carry FOS and-to a lesser extent-FOSB rearrangements suggesting that these tumours are closely related. The presence of these rearrangements could help discriminate these entities from other lesions with woven bone deposition. Osteosarcoma is a malignant bone forming tumour for which different histological subtypes are recognised. High-grade osteosarcoma is the prototype of a complex karyotype tumour, and extensive research exploring its molecular background has identified phenomena like chromothripsis and kataegis and some recurrent alterations. Due to lack of specificity, this has not led to a valuable novel diagnostic marker so far. Nevertheless, these studies have also pointed towards potential targetable drivers of which the therapeutic merit remains to be further explored.


Subject(s)
Bone Neoplasms/pathology , Osteoblastoma/pathology , Osteoma, Osteoid/pathology , Osteosarcoma/pathology , Bone Neoplasms/genetics , Gene Rearrangement , Genes, p53 , Genetic Predisposition to Disease , Humans , Osteoblastoma/genetics , Osteoma/genetics , Osteoma/pathology , Osteoma, Osteoid/genetics , Retinoblastoma Protein/genetics
10.
Genet Test Mol Biomarkers ; 23(11): 807-814, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31638427

ABSTRACT

Background: The gene programmed cell death 5 (PDCD5) has recently been characterized as a tumor suppressor gene and is believed to be an important prognostic cancer marker; it is frequently involved in neoplastic transformation and apoptosis of tumor cells. Several studies have demonstrated a decrease or loss of expression of PDCD5 in certain tumors. However, the relevance of PDCD5 expression in human osteoclastoma and its clinicopathological significance have not been extensively studied. Methods: The aim of this study was to explore the relative transcriptional and translational expression levels of PDCD5 in 79 osteoclastoma samples using multi-modal methods of analysis. Results: Our findings showed that 52% (15/29) of osteoclastoma cases exhibited reduced PDCD5 expression at the transcriptional level, and 56% (44/79) exhibited lower PDCD5 expression at the protein level, when compared with nontumor tissue. In addition, the statistical significance of the altered PDCD5 protein expression was examined using the Campanacci grading system for osteoclastoma. More importantly, the decreased expression at the translational level was observed to have a negative association with the Ki-67 staining index. Conclusion: Based on these findings, abnormal PDCD5 expression might be an important biomarker in human osteoclastoma and may contribute to tumor progression and malignant cell proliferation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Giant Cell Tumor of Bone/genetics , Neoplasm Proteins/genetics , Adult , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , China , Female , Gene Expression Regulation, Neoplastic/genetics , Giant Cell Tumor of Bone/metabolism , Humans , Male , Middle Aged , Neoplasm Proteins/metabolism , Osteoblastoma/genetics , Transcriptome/genetics
11.
Am J Surg Pathol ; 43(12): 1661-1667, 2019 12.
Article in English | MEDLINE | ID: mdl-31490237

ABSTRACT

Osteoblastoma and osteoid osteoma together are the most frequent benign bone-forming tumor, arbitrarily separated by size. In some instances, it can be difficult to differentiate osteoblastoma from osteosarcoma. Following our recent description of FOS gene rearrangement in these tumors, the aim of this study is to evaluate the value of immunohistochemistry in osteoid osteoma, osteoblastoma, and osteosarcoma for diagnostic purposes. A total of 337 cases were tested with antibodies against c-FOS: 84 osteoblastomas, 33 osteoid osteomas, 215 osteosarcomas, and 5 samples of reactive new bone formation. In all, 83% of osteoblastomas and 73% of osteoid osteoma showed significant expression of c-FOS in the osteoblastic tumor cell component. Of the osteosarcomas, 14% showed c-FOS expression, usually focal, and in areas with severe morphologic atypia which were unequivocally malignant: 4% showed more conspicuous expression, but these were negative for FOS gene rearrangement. We conclude that c-FOS immunoreactivity is present in the vast majority of osteoblastoma/osteoid osteoma, whereas its expression is usually focal or patchy, in no more than 14% of osteosarcoma biopsies. Therefore, any bone-forming tumor cases with worrying histologic features would benefit from fluorescence in situ hybridization analysis for FOS gene rearrangement. Our findings highlight the importance of undertaking a thorough assessment of expression patterns of antibodies in the light of morphologic, clinical, and radiologic features.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/chemistry , Osteoblastoma/chemistry , Osteoma, Osteoid/chemistry , Proto-Oncogene Proteins c-fos/analysis , Adolescent , Adult , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Child , Child, Preschool , Diagnosis, Differential , England , Female , Gene Rearrangement , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Osteoblastoma/genetics , Osteoblastoma/pathology , Osteoma, Osteoid/genetics , Osteoma, Osteoid/pathology , Predictive Value of Tests , Proto-Oncogene Proteins c-fos/genetics , Switzerland , Young Adult
12.
Cancer Genomics Proteomics ; 16(5): 361-368, 2019.
Article in English | MEDLINE | ID: mdl-31467230

ABSTRACT

BACKGROUND/AIM: Epithelioid osteoblastoma is a rare benign tumor of the bone. Its pathogenesis is unknown and little is known regarding its genetic features. MATERIALS AND METHODS: Cytogenetic, RNA sequencing, reverse transcription polymerase chain reaction (RT-PCR), genomic PCR, and Sanger sequencing analyses were performed on an epithelioid osteoblastoma. RESULTS: G-banding analysis of short-term cultured tumor cells yielded a normal male karyotype in all examined metaphases. RNA sequencing detected a fusion of COL1A1 from 17q21 with FYN from 6q21. Both RT-PCR and genomic PCR together with Sanger sequencing verified the presence of a COL1A1-FYN fusion gene. In the COL1A1-FYN chimeric transcript, exon 43 of COL1A1 was fused to exon 2 of FYN. The genomic junction occurred in introns 43 and 1 of COL1A1 and FYN, respectively. CONCLUSION: A COL1A1-FYN fusion gene was found in an epithelioid osteoblastoma resulting in deregulation of FYN. Whether COL1A1-FYN represents a consistent genetic feature of epithelioid osteoblastomas, remains to be seen.


Subject(s)
Oncogene Proteins, Fusion/genetics , Osteoblastoma/genetics , Child , Humans , Male , Osteoblastoma/pathology
13.
Nat Commun ; 9(1): 2150, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29858576

ABSTRACT

The transcription factor FOS has long been implicated in the pathogenesis of bone tumours, following the discovery that the viral homologue, v-fos, caused osteosarcoma in laboratory mice. However, mutations of FOS have not been found in human bone-forming tumours. Here, we report recurrent rearrangement of FOS and its paralogue, FOSB, in the most common benign tumours of bone, osteoblastoma and osteoid osteoma. Combining whole-genome DNA and RNA sequences, we find rearrangement of FOS in five tumours and of FOSB in one tumour. Extending our findings into a cohort of 55 cases, using FISH and immunohistochemistry, provide evidence of ubiquitous mutation of FOS or FOSB in osteoblastoma and osteoid osteoma. Overall, our findings reveal a human bone tumour defined by mutations of FOS and FOSB.


Subject(s)
Bone Neoplasms/genetics , Osteoblastoma/genetics , Proto-Oncogene Proteins c-fos/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Bone Neoplasms/diagnosis , Bone Neoplasms/metabolism , Child , Child, Preschool , Female , Gene Rearrangement , Humans , Male , Mice , Middle Aged , Mutation , Osteoblastoma/diagnosis , Osteoblastoma/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Whole Genome Sequencing/methods , Young Adult
14.
Oncotarget ; 8(16): 26424-26433, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460435

ABSTRACT

Forced-activation of AMP-activated protein kinase (AMPK) can possibly inhibit osteoblastoma cells. Here, we aim to provoke AMPK activation via microRNA silencing its phosphatase Ppm1e (protein phosphatase Mg2+/Mn2+-dependent 1e). We showed that microRNA-135b-5p ("miR-135b-5p"), the anti-Ppm1e microRNA, was significantly downregulated in human osteoblastoma tissues. It was correlated with Ppm1e upregulation and AMPKα1 de-phosphorylation. Forced-expression of miR-135b-5p in human osteoblastoma cells (MG-63 and U2OS lines) silenced Ppm1e, and induced a profound AMPKα1 phosphorylation (at Thr-172). Osteoblastoma cell proliferation was inhibited after miR-135b-5p expression. Intriguingly, Ppm1e shRNA knockdown similarly induced AMPKα1 phosphorylation, causing osteoblastoma cell proliferation. Reversely, AMPKα1 shRNA knockdown or dominant negative mutation almost abolished miR-135b-5p's actions in osteoblastoma cells. Further in vivo studies demonstrated that U2OS tumor growth in mice was dramatically inhibited after expressing miR-135b-5p or Ppm1e shRNA. Together, our results suggest that miR-135b-induced Ppm1e silence induces AMPK activation to inhibit osteoblastoma cell proliferation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Gene Silencing , MicroRNAs/genetics , Osteoblastoma/genetics , Osteoblastoma/metabolism , Protein Phosphatase 2C/genetics , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mutation , Osteoblastoma/pathology , Phosphorylation , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
15.
Mol Med Rep ; 12(1): 1157-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25815525

ABSTRACT

The aim of the present study was to identify biomarkers in osteosarcoma (OS) cell serum by antibody microarray profiling, which may be used for OS diagnosis and therapy. An antibody microarray was used to detect the expression levels of cytokines in serum samples from 20 patients with OS and 20 healthy individuals. Significantly expressed cytokines in OS serum were selected when P<0.05 and fold change >2. An enzyme-linked immunosorbent assay (ELISA) was used to validate the antibody microarray results. Finally, classification accuracy was calculated by cluster analysis. Twenty one cytokines were significantly upregulated in OS cell serum samples compared with control samples. Expression of interleukin-6, monocyte chemoattractant protein-1, tumor growth factor-ß, growth-related oncogene, hepatocyte growth factor, chemokine ligand 16, Endoglin, matrix metalloproteinase-9 and platelet-derived growth factor-AA was validated by ELISAs. OS serum samples and control samples were distinguished by significantly expressed cytokines with an accuracy of 95%. The results demonstrated that expressed cytokines identified by antibody microarray may be used as biomarkers for OS diagnosis and therapy.


Subject(s)
Biomarkers, Tumor/blood , Bone Neoplasms/blood , Chondroblastoma/blood , Cytokines/blood , Osteoblastoma/blood , Adolescent , Adult , Antibodies/chemistry , Biomarkers, Tumor/genetics , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Case-Control Studies , Chondroblastoma/diagnosis , Chondroblastoma/genetics , Chondroblastoma/pathology , Cluster Analysis , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Humans , Male , Osteoblastoma/diagnosis , Osteoblastoma/genetics , Osteoblastoma/pathology , Protein Array Analysis/standards , Sensitivity and Specificity
16.
PLoS One ; 8(11): e80725, 2013.
Article in English | MEDLINE | ID: mdl-24236197

ABSTRACT

Osteoblastoma is a bone forming tumor with histological features highly similar to osteoid osteoma; the discrimination between the tumor types is based on size and growth pattern. The vast majority of osteoblastomas are benign but there is a group of so-called aggressive osteoblastomas that can be diagnostically challenging at the histopathological level. The genetic aberrations required for osteoblastoma development are not known and no genetic difference between conventional and aggressive osteoblastoma has been reported. In order to identify recurrent genomic aberrations of importance for tumor development we applied cytogenetic and/or SNP array analyses on nine conventional and two aggressive osteoblastomas. The conventional osteoblastomas showed few or no acquired genetic aberrations while the aggressive tumors displayed heavily rearranged genomes. In one of the aggressive osteoblastomas, three neighboring regions in chromosome band 22q12 were homozygously deleted. Hemizygous deletions of these regions were found in two additional cases, one aggressive and one conventional. In total, 10 genes were recurrently and homozygously lost in osteoblastoma. Four of them are functionally involved in regulating osteogenesis and/or tumorigenesis. MN1 and NF2 have previously been implicated in the development of leukemia and solid tumors, and ZNRF3 and KREMEN1 are inhibitors of the Wnt/beta-catenin signaling pathway. In line with deletions of the latter two genes, high beta-catenin protein expression has previously been reported in osteoblastoma and aberrations affecting the Wnt/beta-catenin pathway have been found in other bone lesions, including osteoma and osteosarcoma.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 22 , Osteoblastoma/genetics , Osteoblastoma/metabolism , Wnt Signaling Pathway , Adolescent , Adult , Child , Cluster Analysis , DNA Copy Number Variations , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Young Adult
17.
Tumour Biol ; 33(5): 1639-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22623071

ABSTRACT

Expression of the p63 tumor suppressor protein has been reported in the mononuclear stromal cells of giant cell tumor of the bone, which may represent osteoblast-precursor cells. Only a limited number of osteoblastic tumors have been studied for p63 expression thus far. We therefore examined whether p63 may serve as a marker for osteoblastic differentiation in osteosarcomas or as a differential diagnostic marker to distinguish osteoblastoma from osteosarcoma. Immunohistochemical stains for p63 were performed on a tissue microarray containing 71 chemotherapy naïve biopsy samples of osteosarcoma, 21 whole sections of osteosarcoma, and 8 osteoblastomas. Nuclear p63 was detected in seven of eight osteoblastomas but was restricted to stromal cells within primitive, immature-appearing areas of osteoid deposition. Although only 7 of 71 (10 %) biopsy samples of osteosarcoma represented on the tissue microarray were positive for p63, 7 of 21 (33 %) osteosarcomas were positive when whole tissue sections were evaluated. Although p63 is detected in most osteoblastomas, it is also observed in a significant subset of osteosarcomas, severely limiting its utility in distinguishing between benign and malignant osteoblastic tumors. The relatively low prevalence of p63 expression in osteosarcoma would also seem to preclude its use as a marker of osteoblastic differentiation in skeletal sarcomas.


Subject(s)
Bone Neoplasms/metabolism , Osteoblastoma/metabolism , Osteosarcoma/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adolescent , Adult , Biomarkers, Tumor , Bone Neoplasms/genetics , Child , Female , Humans , Male , Middle Aged , Osteoblastoma/genetics , Osteosarcoma/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Young Adult
18.
Semin Diagn Pathol ; 28(1): 73-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21675379

ABSTRACT

Benign bone tumors frequently pose a diagnostic challenge for general surgical pathologists. Accurate pathologic diagnosis requires careful clinical and radiological correlation. The most significant recent advances in some benign bone tumors have occurred at the molecular and cytogenetic level. The detection of clonal chromosomal aberrations, various specific molecular genetic events, and the description of the bone cell signaling pathways in the field of osteoimmunology have provided a better understanding of the pathophysiology of certain tumors and an important aid in the diagnostic workup and differential diagnosis of some bone lesions demonstrating overlapping clinical and pathologic features. Future directions include prognostic and therapeutic applications of these findings. Newer less invasive therapeutic techniques and medical management have been developed for the treatment of certain benign bone tumors.


Subject(s)
Bone Neoplasms/diagnosis , Pathology, Surgical/methods , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Chondroblastoma/diagnosis , Chondroblastoma/genetics , Chondroma/diagnosis , Chondroma/genetics , Chromosome Aberrations , Female , Fibrous Dysplasia of Bone/diagnosis , Fibrous Dysplasia of Bone/genetics , Giant Cell Tumor of Bone/diagnosis , Giant Cell Tumor of Bone/genetics , Humans , Male , Notochord/pathology , Osteoblastoma/diagnosis , Osteoblastoma/genetics , Osteochondroma/diagnosis , Osteochondroma/genetics , Osteoma, Osteoid/diagnosis , Osteoma, Osteoid/genetics , Radiography
19.
Hum Pathol ; 41(12): 1788-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21078438

ABSTRACT

Recent progress in skeletal molecular biology has led to the clarification of the transcriptional mechanisms of chondroblastic and osteoblastic lineage differentiation. Three master transcription factors-Sox9, Runx2, and Osterix-were shown to play an essential role in determining the skeletal progenitor cells' fate. The present study evaluates the expression of these factors in 4 types of benign bone tumors-chondromyxoid fibroma, chondroblastoma, osteoid osteoma, and osteoblastoma-using immunohistochemistry and tissue microarrays. Osteoid osteoma and osteoblastoma showed strong nuclear expression of Osterix and Runx2. In contrast, only a few chondroblastomas showed positive nuclear expression of Osterix. Strong nuclear expression of Sox9 was detected in all chondroblastomas, whereas nearly half of the osteoblastomas showed focal weak cytoplasmic expression of Sox9.


Subject(s)
Bone Neoplasms/genetics , Cartilage/growth & development , Gene Regulatory Networks , Neoplasms, Connective Tissue/genetics , Osteogenesis/genetics , Transcription Factors/genetics , Adolescent , Adult , Aged , Bone Neoplasms/pathology , Cartilage/pathology , Child , Chondroblastoma/genetics , Chondroblastoma/metabolism , Chondroblastoma/pathology , Chondroma/genetics , Chondroma/metabolism , Chondroma/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Fibroma/genetics , Fibroma/metabolism , Fibroma/pathology , Gene Expression Regulation , Humans , Male , Middle Aged , Neoplasms, Connective Tissue/metabolism , Neoplasms, Connective Tissue/pathology , Osteoblastoma/genetics , Osteoblastoma/metabolism , Osteoblastoma/pathology , Osteoma, Osteoid/genetics , Osteoma, Osteoid/metabolism , Osteoma, Osteoid/pathology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sp7 Transcription Factor , Stem Cells/metabolism , Stem Cells/pathology , Tissue Array Analysis , Transcription Factors/metabolism , Young Adult
20.
Arch Pathol Lab Med ; 134(10): 1460-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20923301

ABSTRACT

Osteoblastoma is a rare benign bone tumor. Although the histologic features in most cases are distinctive, there are various permutations that make the diagnosis challenging. It can mimic a variety of other benign bone tumors, but more importantly, distinguishing it from osteoblastoma-like osteosarcoma can be difficult. In this case report, I describe the clinicopathologic findings for a 13-year-old adolescent boy with T7 spinal osteoblastoma and review salient clinical, radiographic, and pathologic features of osteoblastoma, as well as the differential diagnoses.


Subject(s)
Bone Neoplasms/pathology , Osteoblastoma/pathology , Spinal Neoplasms/pathology , Adolescent , Bone Neoplasms/diagnosis , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/genetics , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Diagnosis, Differential , Humans , Male , Osteoblastoma/diagnosis , Osteoblastoma/diagnostic imaging , Osteoblastoma/genetics , Osteoblasts/diagnostic imaging , Osteoblasts/pathology , Osteoclasts/diagnostic imaging , Osteoclasts/pathology , Spinal Neoplasms/diagnostic imaging , Spinal Neoplasms/genetics , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...