Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hematol Oncol ; 35(3): 385-389, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28906004

ABSTRACT

Inv(16)(p13q22) and t(16;16)(p13;q22) are cytogenetic hallmarks of acute myelomonoblastic leukaemia, most of them associated with abnormal bone marrow eosinophils [acute myeloid leukaemia French-American-British classification M4 with eosinophilia (FAB AML-M4Eo)] and a relatively favourable clinical course. They generate a 5'CBFB-3'MYH11 fusion gene. However, in a few cases, although RT-PCR identified a CBFB-MYH11 transcript, normal karyotype and/or fluorescent in situ hybridization (FISH) analyses using commercially available probes are found. We identified a 32-year-old woman with AML-M4Eo and normal karyotype and FISH results. Using two libraries of Bacterial Artificial Chromosome clones on 16p13 and 16q22, FISH analyses identified an insertion of 16q22 material in band 16p13, generating a CBFB-MYH11 type A transcript. Although very rare, insertions should be searched for in patients with discordant cytological and cytogenetic features because of the therapeutic consequences. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Mutagenesis, Insertional , Oncogene Proteins, Fusion/genetics , Adult , Biopsy , Bone Marrow Examination , Chromosome Breakpoints , Chromosomes, Human, Pair 16 , Female , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Karyotype , Translocation, Genetic
2.
Future Oncol ; 12(16): 1911-28, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27256160

ABSTRACT

The ROS1 gene belongs to the sevenless subfamily of tyrosine kinase insulin receptor genes. A literature review identified a ROS1 fusion in 2.54% of the patients with lung adenocarcinoma and even higher frequencies in spitzoid neoplasms and inflammatory myofibroblastic tumors. At present, 26 genes were found to fuse with ROS1, some of them already known to fuse with RET and ALK. All the fusion proteins retain the ROS1 kinase domain, but rarely its transmembrane domain. Most of the partners have dimerization domains that are retained in the fusion, presumably leading to constitutive ROS1 tyrosine kinase activation. Some partners have transmembrane domains that are retained or not in the chimeric proteins. Therefore, different ROS1 fusions have distinct subcellular localization, suggesting that they may activate different substrates in vivo.


Subject(s)
Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Humans
3.
Blood Cells Mol Dis ; 54(2): 160-3, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25486926

ABSTRACT

Several chromosomal rearrangements involving band 3q26 are known to induce EVI1 overexpression. They include inv(3)(q21q26), t(3;3)(q21;q26), t(3;21)(q26;q22) and t(3;12)(q26;p13). Translocations involving the short arm of chromosome 2 and 3q26 have been reported in more than 50 patients with myeloid disorders. However, although the breakpoints on 2p are scattered over a long segment, their distribution had only been analyzed in 9 patients. We performed fluorescent in situ hybridization with a library of BAC (Bacterial Artificial Chromosome) clones in 4 patients with t(2;3)(p15-23;q26). Our results combined with those of the 9 previously reported patients showed scattering of the breakpoints in 2 regions. A 1.08Mb region in band 2p21 encompassing the MTA3, ZFP36L2 and THADA genes was documented in 5 patients. A second region of 1.83Mb in band 2p16.1 was identified in 8 patients. Four patients showed clustering around the BCL11A gene and the remaining 4 around a long intergenic non-coding RNA, FLJ30838. These regions are characterized by the presence of regulatory sequences (CpG islands and promoters) that could be instrumental in EVI1 overexpression.


Subject(s)
Anemia, Refractory/genetics , Chromosome Breakpoints , DNA-Binding Proteins/genetics , Genetic Heterogeneity , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Adult , Aged , Anemia, Refractory/pathology , Chromosome Inversion , Chromosomes, Artificial, Bacterial , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 3 , Female , Gene Expression , Gene Library , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/pathology , MDS1 and EVI1 Complex Locus Protein , Male , Middle Aged , Translocation, Genetic
4.
Future Oncol ; 11(11): 1675-86, 2015.
Article in English | MEDLINE | ID: mdl-26043219

ABSTRACT

The EVI1 gene, located in chromosomal band 3q26, is a transcription factor that has stem cell-specific expression pattern and is essential for the regulation of self-renewal of hematopoietic stem cells. It is now recognized as one of the dominant oncogenes associated with myeloid leukemia. EVI1 overexpression is associated with minimal to no response to chemotherapy and poor clinical outcome. Several chromosomal rearrangements involving band 3q26 are known to induce EVI1 overexpression. They are mainly found in acute myeloid leukemia and blastic phase of Philadelphia chromosome-positive chronic myeloid leukemia, more rarely in myelodysplastic syndromes. They include inv(3)(q21q26), t(3;3)(q21;q26), t(3;21)(q26;q22), t(3;12)(q26;p13) and t(2;3)(p15-23;q26). However, many other chromosomal rearrangements involving 3q26/EVI1 have been identified. The precise molecular event has not been elucidated in the majority of these chromosomal abnormalities and most gene partners remain unknown.


Subject(s)
DNA-Binding Proteins/genetics , Gene Rearrangement , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Chromosome Breakpoints , Chromosomes, Human, Pair 3 , Gene Expression , Humans , MDS1 and EVI1 Complex Locus Protein
5.
J Assist Reprod Genet ; 32(4): 615-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25711835

ABSTRACT

Teratozoospermia is characterized by the presence of spermatozoa with abnormal morphology over 85 % in sperm. When all the spermatozoa display a unique abnormality, teratozoospermia is said to be monomorphic. Two forms of monomorphic teratozoospermia, representing less than 1 % of male infertility, are recognized: macrozoospermia (also called macrocephalic sperm head syndrome) and globozoospermia (also called round-headed sperm syndrome). Macrozoospermia is defined as the presence of a very high percentage of spermatozoa with enlarged head and multiple flagella. Meiotic segregation studies in 30 males revealed that over 90 % of spermatozoa were aneuploid, mainly diploid. Sperm DNA fragmentation studies performed in a few patients showed an increase in DNA fragmentation index compared to fertile men. Four mutations in the AURKC gene, a key player in meiosis and more particularly in spermatogenesis, have been found to be responsible for macrozoospermia. Globozoospermia is characterized by round-headed spermatozoa with an absent acrosome, an aberrant nuclear membrane and midpiece defects. The rate of aneuploidy of various chromosomes in spermatozoa from 26 globozoospermic men was slightly increased compared to fertile men. However, this increase was of the same order as that commonly found in infertile men with altered sperm parameters. The majority of the studies found that globozoospermic males had a sperm DNA fragmentation index higher than in fertile men. Mutations or deletions in three genes, SPATA16, PICK1 and DPY19L2, have been shown to be responsible for globozoospermia. Identification of the genetic causes of macrozoospermia and globozoospermia should help refine diagnosis and treatment of these patients, avoiding long and painful treatments. Elucidating the molecular causes of these defects is of utmost importance as intracytoplasmic sperm injection (ICSI) is very disappointing in these two pathologies.


Subject(s)
DNA Fragmentation , Infertility, Male/genetics , Mutation , Spermatozoa/pathology , Humans , Infertility, Male/pathology , Male
6.
Future Oncol ; 10(3): 475-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24559452

ABSTRACT

In humans, class I homeobox genes (HOX genes) are distributed in four clusters. Upstream regulators include transcriptional activators and members of the CDX family of transcription factors. HOX genes encode proteins and need cofactor interactions, to increase their specificity and selectivity. HOX genes contribute to the organization and regulation of hematopoiesis by controlling the balance between proliferation and differentiation. Changes in HOX gene expression can be associated with chromosomal rearrangements generating fusion genes, such as those involving MLL and NUP98, or molecular defects, such as mutations in NPM1 and CEBPA for example. Several miRNAs are involved in the control of HOX gene expression and their expression correlates with HOX gene dysregulation. HOX genes dysregulation is a dominant mechanism of leukemic transformation. A better knowledge of their target genes and the mechanisms by which their dysregulated expression contributes to leukemogenesis could lead to the development of new drugs.


Subject(s)
Gene Expression Regulation, Leukemic , Genes, Homeobox , Leukemia, Myeloid, Acute/genetics , Animals , Genes, Neoplasm , Humans , Leukemia, Myeloid, Acute/metabolism , Multigene Family , Nucleophosmin , Oncogene Proteins, Fusion/genetics
7.
Acta Cytol ; 58(1): 103-7, 2014.
Article in English | MEDLINE | ID: mdl-24281566

ABSTRACT

BACKGROUND: Primary anaplastic large-cell lymphoma (ALCL) occurring in women with breast implants is very rare. It is usually described as tumor cells infiltrating the periprosthetic capsule. These are most often revealed by a periprosthetic recurrent isolated effusion (seroma cavity), occurring late after implantation of the prosthesis. ALCL is more rarely a tumor or periprosthetic capsular contracture. CASE: We report a 66-year-old woman, initially diagnosed by cytological examination of breast effusion, in whom ALCL appeared two and a half months after the removal of a ruptured implant. Repeated biopsies of the periprosthetic capsule performed in parallel showed fibrous tissue, without tumor proliferation. Only meticulous histological examination of the total capsulectomy identified tumor cells as a thin and discontinuous layer along the inner surface of the capsule without capsular invasion. CONCLUSION: Awareness of the histological pattern of this new clinical entity is important. A total capsulectomy with a good sampling for microscopic examination should be conducted for any suspicion of breast implant-associated ALCL. Cytology-histology correlation is essential.


Subject(s)
Breast Implants/adverse effects , Breast Neoplasms/pathology , Lymphoma, Large-Cell, Anaplastic/pathology , Neoplasms, Second Primary/pathology , Adenocarcinoma/surgery , Aged , Breast Neoplasms/etiology , Female , Humans , Lymphoma, Large-Cell, Anaplastic/etiology , Neoplasms, Second Primary/etiology , Prosthesis Failure
8.
J Neurooncol ; 109(2): 405-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22825724

ABSTRACT

Oligodendroglial tumors (ODTs) are primary tumors of the central nervous system that show recurrent codeletion of whole chromosome arms 1p and 19q. Non-1p/19q-deleted high-grade ODTs can present other genetic aberrations, CDKN2A deletion (9p21.3), EGFR amplification (7p11.2) and/or chromosome 10 loss, which are associated with a poor prognosis. The identification of these abnormalities allowed drafting a histo-molecular classification. The aim of this study was to precisely identify, using array CGH, the genomic hallmarks of these tumors, particularly those that are not deleted on 1p/19q. We studied 14 formalin-fixed paraffin-embedded high-grade ODTs using pangenomic oligonucleotide array CGH with an average resolution of 22.3 kb. The 1p/19q codeletion was found in five anaplastic oligodendrogliomas. The three genomic aberrations carrying a poor prognosis were found, most often associated, in five out of nine tumors not deleted on 1p/19q. In addition, four recurrent copy number alterations, involving genes that participate to cell growth and cycle, were found to be strongly associated in five tumors not deleted on 1p/19q: gain or amplification at 1q32.1 (MDM4, PIK3C2B genes), 12q14.1 (CDK4 gene), 12q14.3-q15 (MDM2 gene) and homozygous deletion at 22q13.1 (APOBEC3B gene). MDM2, MDM4, CDK4 and PIK3C2B are known for potentially being amplified or overexpressed in high-grade gliomas. However, the involvement of APOBEC3B, coding for mRNA edition enzyme, is described here for the first time. Our results show a strong association between these four alterations. Therefore, this can open a perspective for a novel subgroup in high-grade ODTs not deleted on 1p/19q.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Loss of Heterozygosity , Oligodendroglioma/genetics , Cell Cycle Proteins , Chromosome Aberrations , Class II Phosphatidylinositol 3-Kinases , Cyclin-Dependent Kinase 4/genetics , Cytidine Deaminase/genetics , Female , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Minor Histocompatibility Antigens , Nuclear Proteins/genetics , Oligodendroglioma/diagnosis , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics
9.
Ann Pathol ; 32(4): 254-8, 2012 Aug.
Article in French | MEDLINE | ID: mdl-23010398

ABSTRACT

Malignant melanoma is a relatively rare but potentially aggressive tumor in children and adolescents. We report the case of a metastatic malignant melanoma in a 17-year-old girl, first diagnosed on cytological features of a fine-needle lymph node aspiration and then histologically confirmed by both examination of the metastatic adenopathy and a clinically harmless skin lesion of the scalp, which harbored focal microscopic pattern of melanoma. A fluorescent in situ hybridization study revealed that both metastatic and primary cutaneous tumours contained the same and pejorative chromosomal aberration consisting in CCND1 amplification (11q13). This observation raises actual limits and challenges in the fields of diagnosis and treatment of fast-killing melanomas.


Subject(s)
Head and Neck Neoplasms/diagnosis , Melanoma/diagnosis , Neoplasms, Second Primary/diagnosis , Scalp/pathology , Skin Neoplasms/diagnosis , Adolescent , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Back Pain/etiology , Combined Modality Therapy , Cyclin D1/genetics , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Fatal Outcome , Female , Gene Amplification , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/surgery , Humans , Immunotherapy , In Situ Hybridization, Fluorescence , Ipilimumab , Lymphatic Metastasis/diagnosis , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Melanoma/secondary , Melanoma/surgery , Nausea/etiology , Neoplasm Proteins/genetics , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Nevus/pathology , Osteolysis/etiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Weight Loss
13.
Am J Med Genet A ; 155A(9): 2281-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21834034

ABSTRACT

We report here three children with a der(11)t(11;16), two sibs (patients 1 and 2) having inherited a recombinant chromosome from a maternal t(11;16)(q24.3;q23.2) and a third unrelated child with a de novo der(11)t(11;16)(q25;q22.1), leading to partial monosomy 11q and trisomy 16q. Fluorescent in situ hybridization (FISH) using bacterial artificial chromosome (BAC) clones and array-CGH were performed to determine the breakpoints involved in the familial and the de novo rearrangements. The partial 11 monosomy extended from 11q24.3 to 11qter and measured 6.17-6.21 Mb in Patients 1 and 2 while the size of the partial 11q25->qter monosomy was estimated at 1.97-2.11 Mb for Patient 3. The partial 16 trisomy extended from 16q23.2 to 16qter and measured 8.93-8.95 Mb in Patients 1 and 2 while the size of the partial 16q22.1->qter trisomy was 20.82 Mb for Patient 3. Intraventricular hemorrhage and transitional thrombocytopenia were found in both sibs but not in the third patient. The FLI1 gene, which is the most relevant gene for thrombocytopenia in Jacobsen syndrome, was neither deleted in family A nor in Patient 3. We suggest that a positional effect could affect the FLI1 expression for these two sibs. Deafness of our three patients confirmed the association of this anomaly to 11q monosomy and tended to confirm the hypothetic role of DFNB20 in Jacobsen syndrome hearing loss. Both sibs shared most of the features commonly observed in Jacobsen syndrome, but not the third patient. This confirmed that terminal 11q trisomy spanning 1 to 1.97-2.11 Mb is not associated with a typical Jacobsen syndrome.


Subject(s)
Chromosome Breakage , Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , Jacobsen Distal 11q Deletion Syndrome/genetics , Trisomy/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 16/genetics , Deafness/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotype , Male , Proto-Oncogene Protein c-fli-1/genetics , Siblings , Translocation, Genetic
14.
J Biomed Biotechnol ; 2011: 329471, 2011.
Article in English | MEDLINE | ID: mdl-21274439

ABSTRACT

The development of the bacterial artificial chromosome (BAC) system was driven in part by the human genome project in order to construct genomic DNA libraries and physical maps for genomic sequencing. The availability of BAC clones has become a valuable tool for identifying cancer genes. We report here our experience in identifying genes located at breakpoints of chromosomal rearrangements and in defining the size and boundaries of deletions in hematological diseases. The methodology used in our laboratory consists of a three-step approach using conventional cytogenetics followed by FISH with commercial probes, then BAC clones. One limitation to the BAC system is that it can only accommodate inserts of up to 300 kb. As a consequence, analyzing the extent of deletions requires a large amount of material. Array comparative genomic hybridization (array-CGH) using a BAC/PAC system can be an alternative. However, this technique has limitations also, and it cannot be used to identify candidate genes at breakpoints of chromosomal rearrangements such as translocations, insertions, and inversions.


Subject(s)
Chromosomes, Artificial, Bacterial , Cytogenetic Analysis/methods , Leukemia/genetics , Biomedical Research , France , Humans
16.
Eur J Haematol ; 86(5): 361-71, 2011 May.
Article in English | MEDLINE | ID: mdl-21435002

ABSTRACT

Chromosomal rearrangements involving the ABL1 gene, leading to a BCR-ABL1 fusion gene, have been mainly associated with chronic myeloid leukemia and B-cell acute lymphoblastic leukemia (ALL). At present, six other genes have been shown to fuse to ABL1. The kinase domain of ABL1 is retained in all chimeric proteins that are also composed of the N-terminal part of the partner protein that often includes a coiled-coil or a helix-loop-helix domain. These latter domains allow oligomerization of the protein that is required for tyrosine kinase activation, cytoskeletal localization, and neoplastic transformation. Fusion genes that have a break in intron 1 or 2 (BCR-ABL1, ETV6-ABL1, ZMIZ1-ABL1, EML1-ABL1, and NUP214-ABL1) have transforming activity, although NUP214-ABL1 requires amplification to be efficient. The NUP214-ABL1 gene is the second most prevalent fusion gene involving ABL1 in malignant hemopathies, with a frequency of 5% in T-cell ALL. Both fusion genes (SFPQ-ABL1 and RCSD1-ABL1) characterized by a break in intron 4 of ABL1 are associated with B-cell ALL, as the chimeric proteins lacked the SH2 domain of ABL1. Screening for ABL1 chimeric genes could be performed in patients with ALL, more particularly in those with T-cell ALL because ABL1 modulates T-cell development and plays a role in cytoskeletal remodeling processes in T cells.


Subject(s)
Genes, abl , Hematologic Neoplasms/genetics , Oncogene Fusion , Cell Transformation, Neoplastic/genetics , Hematologic Neoplasms/chemistry , Humans , Intracellular Signaling Peptides and Proteins/genetics , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Oncogene Proteins, Fusion/genetics , PTB-Associated Splicing Factor , Protein-Tyrosine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics
17.
Future Oncol ; 7(1): 77-91, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21174539

ABSTRACT

The RUNX1 gene, located in chromosome 21q22, is crucial for the establishment of definitive hematopoiesis and the generation of hematopoietic stem cells in the embryo. It contains a 'Runt homology domain' as well as transcription activation and inhibition domains. RUNX1 can act as activator or repressor of target gene expression depending upon the large number of transcription factors, coactivators and corepressors that interact with it. Translocations involving chromosomal band 21q22 are regularly identified in leukemia patients. Most of them are associated with a rearrangement of RUNX1. Indeed, at present, 55 partner chromosomal bands have been described but the partner gene has solely been identified in 21 translocations at the molecular level. All the translocations that retain Runt homology domains but remove the transcription activation domain have a leukemogenic effect by acting as dominant negative inhibitors of wild-type RUNX1 in transcription activation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Fusion , Hematologic Neoplasms/genetics , Translocation, Genetic , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Rearrangement , Genetic Association Studies , Humans
20.
Blood Cells Mol Dis ; 44(4): 268-74, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20206559

ABSTRACT

Chromosomal rearrangements involving the MLL gene have been associated with many different types of hematological malignancies. Most of them are easily recognized by conventional cytogenetics. However, in some cases, complex, unusual or cryptic rearrangements make the MLL involvement difficult or impossible to be detected by conventional cytogenetics. Fluorescent in situ hybridization with a panel of probes coupled with long distance inverse-PCR was used to identify chromosomal rearrangements involving the MLL gene. Seven unusual chromosomal rearrangements were identified, including two complex translocations, three insertions of material of chromosome 11 in another chromosome and one insertion of chromosome material into the MLL gene. Conventional cytogenetics showed three patients to have a deletion of 11q; one had an unexpected t(6;11)(q27;q23) whereas the other two patients had also an insertion of MLL material in another chromosome. Concurrent 3' deletion in the MLL rearrangement was observed in two patients. We recommend a systematic approach to be used in all cases of acute leukemia starting with FISH analyses using a commercially available MLL split signal probe. Should an abnormality be discovered, the analysis has to be completed by further molecular cytogenetic and genomic PCR methods in order to unravel the recombination mechanism.


Subject(s)
Chromosome Aberrations , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Acute Disease , Adenocarcinoma , Adult , Aged , Blast Crisis/genetics , Child, Preschool , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Duodenal Neoplasms , Female , Histone-Lysine N-Methyltransferase , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Leukemia/pathology , Leukemia, Monocytic, Acute/congenital , Leukemia, Monocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Male , Mutagenesis, Insertional , Neoplasms, Second Primary/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prostatic Neoplasms , Sequence Deletion , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL