Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 5 de 5
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Appl Immunohistochem Mol Morphol ; 25(3): 168-177, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-26808135

RÉSUMÉ

INTRODUCTION: Sarcomas are heterogeneous, and their treatment and prognosis are driven by the morphologic subtype and the clinical stage. Classic cytogenetics and fluorescence in situ hybridization (FISH) analysis play an important role in their diagnostic work up. MATERIALS AND METHODS: Forty-six cases of soft-tissue sarcoma were reviewed that underwent karyotyping and simultaneous FISH analysis at initial diagnosis. They included 10 dedifferentiated liposarcomas, 10 myxoid liposarcomas, and 14 synovial sarcomas. Six tumors were investigated for EWSR1 rearrangement. Six high-grade miscellaneous sarcomas were also examined. RESULTS: The dedifferentiated liposarcoma had complex karyotypes and MDM2 amplification by FISH, and of these, 5 tumors with myxoid changes also had complex signals for DDIT3. All but 4 myxoid liposarcomas had complex karyotypes, in addition to the characteristic translocation. FISH analysis displayed DD1T3 rearrangement. All synovial sarcomas except 1 recurrence had a t(X;18) translocation by karyotyping and FISH. The EWSR1 rearrangement was present in all extraskeletal myxoid chondrosarcomas, angiomatoid fibrous histiocytoma, atypical Ewing sarcoma, and a clear-cell sarcoma, all of which had characteristic karyotypes. Seven high-grade sarcomas had no specific karyotype or rearrangements for DDIT3, SS18, and EWSR1 by FISH. CONCLUSIONS: There is good correlation between karyotyping and FISH. Complex FISH signals found in dedifferentiated liposarcomas may be related to an increased chromosome 12 copy number and ploidy. Karyotyping is an important baseline standard for the quality assurance of newly developed FISH probes. It also provides a global view of chromosomal changes and the opportunity to investigate the role of other genetic alterations and potential therapeutic targets.


Sujet(s)
Tumeurs du tissu adipeux/anatomopathologie , Sarcome synovial/anatomopathologie , Sarcomes/génétique , Humains , Hybridation fluorescente in situ , Tumeurs du tissu adipeux/génétique , Études rétrospectives , Sarcomes/anatomopathologie , Sarcome synovial/génétique
2.
Genes Chromosomes Cancer ; 55(9): 694-709, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27123539

RÉSUMÉ

Oral squamous cell carcinoma (OSCC) is a serious public health problem caused primarily by smoking and alcohol consumption or human papillomavirus. The cancer stem cell (CSC) theory posits that CSCs show unique characteristics, including self-renewal and therapeutic resistance. Examining biomarkers and other features of CSCs is critical to better understanding their biology. To this end, the results show that cellular SOX2 immunostaining correlates with other CSC biomarkers in OSCC cell lines and marks the rare CSC population. To assess whether CSC division patterns are symmetrical, resulting in two CSC, or asymmetrical, leading to one CSC and one cancer cell, cell size and fluorescence intensity of mitotic cells stained with SOX2 were analyzed. Asymmetrical SOX2 distribution in ≈25% of the mitoses analyzed was detected. Chromosomal instability, some of which is caused by chromosome segregation defects (CSDs), is a feature of cancer cells that leads to altered gene copy numbers. We compare chromosomal instability (as measured by CSDs) between CSCs (SOX2+) and non-CSCs (SOX2-) from the same OSCC cell lines. CSDs were more common in non-CSCs (SOX2-) than CSCs (SOX2+) and in symmetrical CSC (SOX2+) mitotic pairs than asymmetrical CSC (SOX2+/SOX2-) mitotic pairs. CSCs showed fewer and different types of CSDs after ionizing radiation treatment than non-CSCs. Overall, these data are the first to demonstrate both symmetrical and asymmetrical cell divisions with CSDs in OSCC CSC. Further, the results suggest that CSCs may undergo altered behavior, including therapeutic resistance as a result of chromosomal instability due to chromosome segregation defects. © 2016 Wiley Periodicals, Inc.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Carcinome épidermoïde/anatomopathologie , Division cellulaire/génétique , Ségrégation des chromosomes/génétique , Tumeurs de la bouche/anatomopathologie , Cellules souches tumorales/anatomopathologie , Facteurs de transcription SOX-B1/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinome épidermoïde/génétique , Différenciation cellulaire/génétique , Différenciation cellulaire/effets des radiations , Division cellulaire/effets des radiations , Ségrégation des chromosomes/effets des radiations , Technique d'immunofluorescence , Humains , Rayons infrarouges , Tumeurs de la bouche/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des radiations , Facteurs de transcription SOX-B1/métabolisme , Cellules cancéreuses en culture
3.
Genes Chromosomes Cancer ; 53(12): 972-90, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25183546

RÉSUMÉ

Cytogenetic alterations underlie the development of head and neck squamous cell carcinoma (HNSCC), whether tobacco and alcohol use, betel nut chewing, snuff or human papillomavirus (HPV) causes the disease. Many of the molecular genetic aberrations in HNSCC result from these cytogenetic alterations. This review presents a brief introduction to the epidemiology of HNSCC, and discusses the role of HPV in the disease, cytogenetic alterations and their frequencies in HNSCC, their molecular genetic and The Cancer Genome Atlas (TCGA) correlates, prognostic implications, and possible therapeutic considerations. The most frequent cytogenetic alterations in HNSCC are gains of 5p14-15, 8q11-12, and 20q12-13, gains or amplifications of 3q26, 7p11, 8q24, and 11q13, and losses of 3p, 4q35, 5q12, 8p23, 9p21-24, 11q14-23, 13q12-14, 18q23, and 21q22. To understand their effects on tumor cell biology and response to therapy, the cytogenetic findings in HNSCC are increasingly being examined in the context of the biochemical pathways they disrupt. The goal is to minimize morbidity and mortality from HNSCC using cytogenetic abnormalities to identify valuable diagnostic biomarkers for HNSCC, prognostic biomarkers of tumor behavior, recurrence risk, and outcome, and predictive biomarkers of therapeutic response to identify the most efficacious treatment for each individual patient's tumor, all based on a detailed understanding of the next generation biology of HNSCC.


Sujet(s)
Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/génétique , Aberrations des chromosomes , Tumeurs de la tête et du cou/diagnostic , Tumeurs de la tête et du cou/génétique , Marqueurs biologiques tumoraux/génétique , Carcinome épidermoïde/anatomopathologie , Tumeurs de la tête et du cou/anatomopathologie , Humains , Pronostic , Carcinome épidermoïde de la tête et du cou
4.
Genes Chromosomes Cancer ; 53(2): 129-43, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24327542

RÉSUMÉ

Oral squamous cell carcinoma (OSCC), a subset of head and neck squamous cell carcinoma (HNSCC), is the eighth most common cancer in the U.S.. Amplification of chromosomal band 11q13 and its association with poor prognosis has been well established in OSCC. The first step in the breakage-fusion-bridge (BFB) cycle leading to 11q13 amplification involves breakage and loss of distal 11q. Distal 11q loss marked by copy number loss of the ATM gene is observed in 25% of all Cancer Genome Atlas (TCGA) tumors, including 48% of HNSCC. We showed previously that copy number loss of distal 11q is associated with decreased sensitivity (increased resistance) to ionizing radiation (IR) in OSCC cell lines. We hypothesized that this radioresistance phenotype associated with ATM copy number loss results from upregulation of the compensatory ATR-CHEK1 pathway, and that knocking down the ATR-CHEK1 pathway increases the sensitivity to IR of OSCC cells with distal 11q loss. Clonogenic survival assays confirmed the association between reduced sensitivity to IR in OSCC cell lines and distal 11q loss. Gene and protein expression studies revealed upregulation of the ATR-CHEK1 pathway and flow cytometry showed G2 M checkpoint arrest after IR treatment of cell lines with distal 11q loss. Targeted knockdown of the ATR-CHEK1 pathway using CHEK1 or ATR siRNA or a CHEK1 small molecule inhibitor (SMI, PF-00477736) resulted in increased sensitivity of the tumor cells to IR. Our results suggest that distal 11q loss is a useful biomarker in OSCC for radioresistance that can be reversed by ATR-CHEK1 pathway inhibition.


Sujet(s)
Carcinome épidermoïde/génétique , Chromosomes humains de la paire 11/génétique , Tumeurs de la bouche/génétique , Protein kinases/génétique , Radiotolérance , Protéines mutées dans l'ataxie-télangiectasie/génétique , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Carcinome épidermoïde/radiothérapie , Lignée cellulaire tumorale/effets des radiations , Checkpoint kinase 1 , Délétion de segment de chromosome , Ségrégation des chromosomes , Altération de l'ADN , Techniques de knock-down de gènes , Humains , Points de contrôle de la phase M du cycle cellulaire , Tumeurs de la bouche/radiothérapie , Inhibiteurs de protéines kinases/pharmacologie , Protein kinases/métabolisme , Transduction du signal , Régulation positive
5.
Genes Chromosomes Cancer ; 53(1): 25-37, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24142626

RÉSUMÉ

The ATR-CHEK1 pathway is upregulated and overactivated in Ataxia Telangiectasia (AT) cells, which lack functional ATM protein. Loss of ATM in AT confers radiosensitivity, although ATR-CHEK1 pathway overactivation compensates, leads to prolonged G(2) arrest after treatment with ionizing radiation (IR), and partially reverses the radiosensitivity. We observed similar upregulation of the ATR-CHEK1 pathway in a subset of oral squamous cell carcinoma (OSCC) cell lines with ATM loss. In the present study, we report copy number gain, amplification, or translocation of the ATR gene in 8 of 20 OSCC cell lines by FISH; whereas the CHEK1 gene showed copy number loss in 12 of 20 cell lines by FISH. Quantitative PCR showed overexpression of both ATR and CHEK1 in 7 of 11 representative OSCC cell lines. Inhibition of ATR or CHEK1 with their respective siRNAs resulted in increased sensitivity of OSCC cell lines to IR by the colony survival assay. siRNA-mediated ATR or CHEK1 knockdown led to loss of G(2) cell cycle accumulation and an increased sub-G(0) apoptotic cell population by flow cytometric analysis. In conclusion, the ATR-CHEK1 pathway is upregulated in a subset of OSCC with distal 11q loss and loss of the G(1) phase cell cycle checkpoint. The upregulated ATR-CHEK1 pathway appears to protect OSCC cells from mitotic catastrophe by enhancing the G(2) checkpoint. Knockdown of ATR and/or CHEK1 increases the sensitivity of OSCC cells to IR. These findings suggest that inhibition of the upregulated ATR-CHEK1 pathway may enhance the efficacy of ionizing radiation treatment of OSCC.


Sujet(s)
Carcinome épidermoïde/génétique , Tumeurs de la bouche/génétique , Protein kinases/génétique , Protéines mutées dans l'ataxie-télangiectasie/génétique , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Checkpoint kinase 1 , Chromosomes humains de la paire 11/génétique , Chromosomes humains de la paire 3/génétique , Altération de l'ADN/effets des radiations , Points de contrôle de la phase G2 du cycle cellulaire/effets des radiations , Dosage génique , Techniques de knock-down de gènes , Humains , Tumeurs de la bouche/métabolisme , Protein kinases/métabolisme , Radiotolérance , Transduction du signal , Translocation génétique , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE