Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
2.
J Mol Diagn ; 24(9): 1021-1030, 2022 09.
Article En | MEDLINE | ID: mdl-35718095

Targeted therapy in lung cancer requires the assessment of multiple oncogenic driver alterations, including fusion genes. This retrospective study evaluated the Idylla GeneFusion prototype, an automated and ease-of-use (<2 minutes) test, with a short turnaround time (3 hours) to detect fusions involving ALK, ROS1, RET, and NTRK1/2/3 genes and MET exon 14 skipping. This multicenter study (18 centers) included 313 tissue samples from lung cancer patients with 97 ALK, 44 ROS1, 20 RET, and 5 NTRKs fusions, 32 MET exon 14 skipping, and 115 wild-type samples, previously identified with reference methods (RNA-based next-generation sequencing/fluorescence in situ hybridization/quantitative PCR). Valid results were obtained for 306 cases (98%), overall concordance between Idylla and the reference methods was 89% (273/306); overall sensitivity and specificity were 85% (165/193) and 96% (108/113), respectively. Discordances were observed in 28 samples, where Idylla did not detect the alteration identified by the reference methods; and 5 samples where Idylla identified an alteration not detected by the reference methods. All of the ALK-, ROS1-, and RET-specific fusions and MET exon 14 skipping identified by Idylla GeneFusion were confirmed by reference method. To conclude, Idylla GeneFusion is a clinically valuable test that does not require a specific infrastructure, allowing a rapid result. The absence of alteration or the detection of expression imbalance only requires additional testing by orthogonal methods.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Mutation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Retrospective Studies
3.
BMC Cancer ; 19(1): 81, 2019 Jan 17.
Article En | MEDLINE | ID: mdl-30654767

BACKGROUND: Age-related genetic changes in lymphocyte subsets are not currently well documented. BACH2 is a transcription factor that plays an important role in immune-mediated homeostasis by tightly regulating PRDM1 expression in both B-cells and T-cells. BACH2 gene expression is highly sensitive to DNA damage in aged mice. This concept led us to investigate the variation in BACH2 and also PRDM1 expression in major lymphocyte subsets with age. METHODS: Lymphocyte subsets from 60 healthy donors, aged from 20 to 90 years, and 41 untreated chronic lymphocytic leukemia patients were studied. BACH2 and PRDM1 gene expression was analyzed by real-time quantitative PCR. BACH2 gene expression was correlated with its protein expression. Lymphocyte apoptosis was evaluated after intracellular oxidative stress-inducing etoposide treatment of T and B cells. RESULTS: Our analysis shows BACH2 mRNA downregulation with age in healthy donor CD4+, CD8+ T-cells and CD19+ B-cells. Decreased BACH2 expression was also correlated with an age-related reduction in CD8 + CD28+ T-cells. We found a strong correlation between age-related BACH2 downregulation and decreased CD4+ T-cell and CD19+ B-cell apoptosis. PRDM1, as expected, was significantly upregulated in CD4+ T-cells, CD8+ T-cells and CD19+ B-cells, and inversely correlated with BACH2. A comparison of untreated chronic lymphocytic leukemia patients with age-matched healthy donors reveals that BACH2 mRNA expression was further reduced in CD4+ T-cells, CD8+ T-cells and leukemic-B cells. PRDM1 gene expression was consequently significantly upregulated in CD4+ and CD8+ T-cells in chronic lymphocytic leukemia patients but not in their leukemic B-cells. CONCLUSION: Overall, our data suggest that BACH2 and PRDM1 genes are significantly correlated with age in human immune cells and may be involved in immunosenescence.


Aging/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Subsets/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Adult , Aged , Aged, 80 and over , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cellular Senescence/immunology , Down-Regulation/immunology , Female , Healthy Volunteers , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocyte Subsets/immunology , Male , Middle Aged , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , RNA, Messenger/metabolism , Up-Regulation/immunology , Young Adult
4.
Cardiovasc Res ; 112(1): 478-90, 2016 10.
Article En | MEDLINE | ID: mdl-27520736

AIM: Cardiac progenitor cells (CPC) from adult hearts can differentiate to several cell types composing the myocardium but the underlying molecular pathways are poorly characterized. We examined the role of paracrine nitric oxide (NO) in the specification of CPC to the cardiac lineage, particularly through its inhibition of the canonical Wnt/ß-catenin pathway, a critical step preceding cardiac differentiation. METHODS AND RESULTS: Sca1 + CPC from adult mouse hearts were isolated by magnetic-activated cell sorting and clonally expanded. Pharmacologic NO donors increased their expression of cardiac myocyte-specific sarcomeric proteins in a concentration and time-dependent manner. The optimal time window for NO efficacy coincided with up-regulation of CPC expression of Gucy1a3 (coding the alpha1 subunit of guanylyl cyclase). The effect of paracrine NO was reproduced in vitro upon co-culture of CPC with cardiac myocytes expressing a transgenic NOS3 (endothelial nitric oxide synthase) and in vivo upon injection of CPC in infarcted hearts from cardiac-specific NOS3 transgenic mice. In mono- and co-cultures, this effect was abrogated upon inhibition of soluble guanylyl cyclase or nitric oxide synthase, and was lost in CPC genetically deficient in Gucy1a3. Mechanistically, NO inhibits the constitutive activity of the canonical Wnt/ß-catenin in CPC and in cell reporter assays in a guanylyl cyclase-dependent fashion. This was paralleled with decreased expression of ß-catenin and down-regulation of Wnt target genes in CPC and abrogated in CPC with a stabilized, non-inhibitable ß-catenin. CONCLUSIONS: Exogenous or paracrine sources of NO promote the specification towards the myocyte lineage and expression of cardiac sarcomeric proteins of adult CPC. This is contingent upon the expression and activity of the alpha1 subunit of guanylyl cyclase in CPC that is necessary for NO-mediated inhibition of the canonical Wnt/ß-catenin pathway.


Adult Stem Cells/metabolism , Cell Differentiation , Cyclic GMP/metabolism , Myocytes, Cardiac/enzymology , Nitric Oxide/metabolism , Paracrine Communication , Sarcomeres/enzymology , Soluble Guanylyl Cyclase/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adult Stem Cells/drug effects , Animals , Antigens, Ly/metabolism , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Female , Immunomagnetic Separation , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Paracrine Communication/drug effects , Sarcomeres/drug effects , Signal Transduction , Soluble Guanylyl Cyclase/deficiency , Soluble Guanylyl Cyclase/genetics , Time Factors , Transfection , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
5.
Eur J Cancer ; 55: 98-110, 2016 Mar.
Article En | MEDLINE | ID: mdl-26790143

Intrinsic and acquired resistance of metastatic melanoma to (V600E/K)BRAF and/or MEK inhibitors, which is often caused by activation of the PI3K/AKT survival pathway, represents a major clinical challenge. Given that p53 is capable of antagonising PI3K/AKT activation we hypothesised that pharmacological restoration of p53 activity may increase the sensitivity of BRAF-mutant melanoma to MAPK-targeted therapy and eventually delay and/or prevent acquisition of drug resistance. To test this possibility we exposed a panel of vemurafenib-sensitive and resistant (innate and acquired) (V600E/K)BRAF melanomas to a (V600E/K)BRAF inhibitor (vemurafenib) alone or in combination with a direct p53 activator (PRIMA-1(Met)/APR-246). Strikingly, PRIMA-1(Met) synergised with vemurafenib to induce apoptosis and suppress proliferation of (V600E/K)BRAF melanoma cells in vitro and to inhibit tumour growth in vivo. Importantly, this drug combination decreased the viability of both vemurafenib-sensitive and resistant melanoma cells irrespectively of the TP53 status. Notably, p53 reactivation was invariably accompanied by PI3K/AKT pathway inhibition, the activity of which was found as a dominant resistance mechanism to BRAF inhibition in our lines. From all various combinatorial modalities tested, targeting the MAPK and PI3K signalling pathways through p53 reactivation or not, the PRIMA-1(Met)/vemurafenib combination was the most cytotoxic. We conclude that PRIMA-1(Met) through its ability to directly reactivate p53 regardless of the mechanism causing its deactivation, and thereby dampen PI3K signalling, sensitises (V600E/K)BRAF-positive melanoma to BRAF inhibitors.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indoles/pharmacology , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Quinuclidines/pharmacology , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Genetic Predisposition to Disease , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Molecular Targeted Therapy , Mutation , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Tumor Suppressor Protein p53/genetics , Vemurafenib , Xenograft Model Antitumor Assays
6.
Asian Pac J Cancer Prev ; 16(9): 4051-5, 2015.
Article En | MEDLINE | ID: mdl-25987085

The purpose of this study is to assess the effect of consanguinity on breast cancer incidence in Tunisia. We conducted a case-control study to evaluate the involvement of heterozygote and homozygote haplotypes of BRCA1 gene SNPs according to consanguinity among 40 cases of familial breast cancer, 46 cases with sporadic breast cancer and 34 healthy controls. We showed significant difference in consanguinity rate between breast cancer patients versus healthy controls P = 0.001. Distribution of homozygous BRCA1 haplotypes among healthy women versus breast cancer patients was significantly different; p=0.02. Parental consanguinity seems to protect against breast cancer in the Tunisian population.


BRCA1 Protein/genetics , Consanguinity , Haplotypes/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Breast Neoplasms/prevention & control , Case-Control Studies , Female , Follow-Up Studies , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Incidence , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Prospective Studies , Tunisia/epidemiology , Young Adult
7.
Pathol Res Pract ; 210(6): 389-91, 2014 Jun.
Article En | MEDLINE | ID: mdl-24629658

Breast myofibroblastomas are rare benign mesenchymal tumors belonging to the group of stromal breast tumors composed of spindle-shaped cells and characterized by a broad morphologic spectrum. Among the different morphologic variants described, breast MFBs can show smooth muscle cell differentiation in very rare cases. In terms of the genetic abnormalities found in this type of tumor, a deletion of chromosome 13q14 was recently confirmed by FISH in some cases of mammary MFB. In this paper, we report an unusual case of MFB with smooth muscle differentiation showing a deletion of chromosome 13q14.


Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Cell Differentiation , Chromosome Disorders/diagnosis , Forkhead Transcription Factors/genetics , Gene Deletion , Myocytes, Smooth Muscle/pathology , Neoplasms, Muscle Tissue/diagnosis , Aged , Biomarkers, Tumor/analysis , Biopsy , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Chromosome Deletion , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosomes, Human, Pair 13/genetics , Female , Forkhead Box Protein O1 , Genetic Predisposition to Disease , Humans , In Situ Hybridization, Fluorescence , Myocytes, Smooth Muscle/chemistry , Neoplasms, Muscle Tissue/chemistry , Neoplasms, Muscle Tissue/genetics , Neoplasms, Muscle Tissue/pathology , Neoplasms, Muscle Tissue/surgery , Phenotype , Predictive Value of Tests , Ultrasonography, Mammary
8.
J Clin Invest ; 123(7): 2873-92, 2013 Jul.
Article En | MEDLINE | ID: mdl-23778140

CD4⁺ T cells are critical regulators of immune responses, but their functional role in human breast cancer is relatively unknown. The goal of this study was to produce an image of CD4⁺ T cells infiltrating breast tumors using limited ex vivo manipulation to better understand the in vivo differences associated with patient prognosis. We performed comprehensive molecular profiling of infiltrating CD4⁺ T cells isolated from untreated invasive primary tumors and found that the infiltrating T cell subpopulations included follicular helper T (Tfh) cells, which have not previously been found in solid tumors, as well as Th1, Th2, and Th17 effector memory cells and Tregs. T cell signaling pathway alterations included a mixture of activation and suppression characterized by restricted cytokine/chemokine production, which inversely paralleled lymphoid infiltration levels and could be reproduced in activated donor CD4⁺ T cells treated with primary tumor supernatant. A comparison of extensively versus minimally infiltrated tumors showed that CXCL13-producing CD4⁺ Tfh cells distinguish extensive immune infiltrates, principally located in tertiary lymphoid structure germinal centers. An 8-gene Tfh signature, signifying organized antitumor immunity, robustly predicted survival or preoperative response to chemotherapy. Our identification of CD4⁺ Tfh cells in breast cancer suggests that they are an important immune element whose presence in the tumor is a prognostic factor.


Breast Neoplasms/immunology , T-Lymphocytes, Helper-Inducer/immunology , Antigens, CD/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome
10.
Blood ; 114(14): 2969-83, 2009 Oct 01.
Article En | MEDLINE | ID: mdl-19608752

The clonal CD3(-)CD4(+) T-cell population characterizing lymphocytic variant hypereosinophilic syndrome (L-HES) persists for years, with a subgroup of patients ultimately progressing to T lymphoma. The molecular changes associated with the premalignant clone and the emergence of malignant subclones are unknown, precluding the development of targeted therapy for this HES variant. In this study, we used whole genome arrays to examine gene expression in the CD3(-)CD4(+) T cells and found that 850 genes were differentially regulated during chronic disease compared with CD3(+)CD4(+) T cells from healthy donors. Changes in the expression of 349 genes were altered in association with the clinical progression from chronic L-HES to T lymphoma in 1 patient, with 87 of 349 genes representing further changes in genes whose expression was altered in all chronic disease patients (87 of 850). Array analysis after CD2/CD28-mediated activation revealed that the major gene expression changes observed in the CD3(-)CD4(+) T cells do not reflect activation induced alterations but rather pathways involved in T-cell homeostasis, including transforming growth factor-beta signaling, apoptosis, and T-cell maturation, signaling, and migration. Examination of microRNA expression in the CD3(-)CD4(+) T cells from patients with chronic disease identified 23 microRNAs that changed significantly, among which miR-125a further decreased in association with one patient's evolution to T lymphoma.


CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/pathology , Lymphocytes/metabolism , Adolescent , Adult , Female , Flow Cytometry , Gene Expression Profiling , Humans , Hypereosinophilic Syndrome/metabolism , Male , MicroRNAs/physiology , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Young Adult
11.
Acta Orthop Belg ; 74(1): 1-6, 2008 Feb.
Article En | MEDLINE | ID: mdl-18411594

The final diagnosis of a bone tumour comes in many cases like the last piece of a puzzle which requires integration of clinical, imaging and pathological data. However there are situations in which a discrepancy exists between histology and imaging studies and where histology alone cannot be decisive. This paper reviews such situations.


Bone Neoplasms/pathology , Chondroma/pathology , Giant Cell Tumors/pathology , Humans , Osteosarcoma/pathology
12.
Pediatr Blood Cancer ; 50(3): 694-6, 2008 Mar.
Article En | MEDLINE | ID: mdl-16856159

We report an infant with normal neurological development and phenotype who developed bilateral retinoblastoma (RB). This patient, despite lack of dysmorphic features, demonstrated constitutional abnormality of the long arm of chromosome 13 on standard karyotype. We recommend systematic cytogenetic examinations complemented by fluorescent in situ hybridization as second-line screening in all patients suspected for hereditary RB despite negative RB1 molecular screening and normal phenotype.


Chromosome Deletion , Chromosomes, Human, Pair 13/ultrastructure , Chromosomes, Human, Pair 8/ultrastructure , Genes, Retinoblastoma , Neoplasms, Multiple Primary/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Translocation, Genetic/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 8/genetics , Combined Modality Therapy , Etoposide/administration & dosage , Eye Enucleation , False Negative Reactions , Female , Humans , Hyperthermia, Induced , In Situ Hybridization, Fluorescence , Infant , Remission Induction , Retinal Neoplasms/therapy , Retinoblastoma/therapy , Skull/abnormalities , Vincristine/administration & dosage
13.
Hereditas ; 144(5): 191-4, 2007 Nov.
Article En | MEDLINE | ID: mdl-18031353

Herein we describe a unique case of partial proximal 10q trisomy presenting biliary atresia, anal anteposition and cardiac malformation. The 10q duplication was confirmed by G banding on prophase chromosomes. A review of the literature confirmed that the patient displayed characteristic dysmorphic features of the recently defined partial proximal trisomy 10q syndrome and emphasized the interindividual variability of visceral malformations.


Biliary Atresia/genetics , Chromosomes, Human, Pair 10 , Trisomy , Aortic Coarctation/surgery , Biliary Atresia/diagnosis , Chromosome Banding , Humans , Infant, Newborn
14.
Hepatology ; 46(5): 1574-85, 2007 Nov.
Article En | MEDLINE | ID: mdl-17969047

The phenotypic homology of fibroblasts and mesenchymal stem cells (MSCs) has been recently described. Our study investigated the in vitro potential of human skin fibroblasts to differentiate into mesodermal (osteocyte and adipocyte) and endodermal (hepatocyte) cell lineages by comparison with human bone marrow (hBM) MSCs. The endodermal potential of fibroblasts was then explored in vivo in a mouse model of liver injury. Fibroblasts were able to acquire osteocyte and adipocyte phenotypes as assessed by cytochemistry and gene expression analyses. After exposure to a specific differentiation cocktail, these cells presented hepatocyte-like morphology and acquired liver-specific markers on protein and gene expression levels. Furthermore, these fibroblast-derived hepatocyte-like cells (FDHLCs) displayed the ability to store glycogen and synthesize small amounts of urea. By gene expression analysis, we observed that fibroblasts remained in a mesenchymal-epithelial transition state after hepatocyte differentiation. Moreover, FDHLCs lost their hepatocyte-like phenotype after dedifferentiation. In vivo, human fibroblasts infused directly into the liver of hepatectomized severe combined immunodeficient (SCID) mice engrafted in situ and expressed hepatocyte markers (albumin, alpha-fetoprotein, and cytokeratin 18) together with the mesodermal marker fibronectin. Despite lower liver-specific marker expression, the in vitro and in vivo differentiation profile of fibroblasts was comparable to that of mesenchymal-derived hepatocyte-like cells (MDHLCs). In conclusion, our work demonstrates that human skin fibroblasts are able to display mesodermal and endodermal differentiation capacities and provides arguments that these cells share MSCs features both on the phenotypic and functional levels.


Cell Differentiation/physiology , Connective Tissue Cells/cytology , Epithelial Cells/cytology , Fibroblasts/cytology , Skin/cytology , Actins/metabolism , Adolescent , Adult , Cells, Cultured , Child , Connective Tissue Cells/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype , Snail Family Transcription Factors , Transcription Factors/metabolism
15.
Gastroenterology ; 130(4): 1317-23, 2006 Apr.
Article En | MEDLINE | ID: mdl-16618422

BACKGROUND & AIMS: Donor cell engraftment with expression of enzyme activity is the goal of liver cell transplantation for inborn errors of liver metabolism with a view to achieving sustained metabolic control. METHODS: Sequential hepatic cell transplantations using male and female cells were performed in a 3.5-year-old girl with argininosuccinate lyase deficiency over a period of 5 months. Beside clinical, psychomotor, and metabolic follow-up, engraftment was analyzed in repeated liver biopsies (2.5, 5, 8, and 12 months after first infusion) by fluorescence in situ hybridization for the Y-chromosome and by measurement of tissue enzyme activity. RESULTS: Metabolic control was achieved together with psychomotor catch-up, changing the clinical phenotype from a severe neonatal one to a moderate late-onset type. The child was no longer hospitalized and was able to attend normal school. Sustained engraftment of male donor liver cells was shown in repeated biopsies, reaching 19% at 8 months and 12.5% at the 12-month follow-up. XXYY tetraploid donor cells were mainly detected during the infusion period (2.5- and 5-month biopsies), whereas in the follow-up 8-month and 1-year biopsies, diploid donor cell subpopulations had become dominant. Moreover, argininosuccinate lyase activity, originally absent, became measurable in 2 different biopsy samples at 8 months, reaching 3% of control activity, indicating in situ metabolic effect and supporting the clinical evolution to a moderate form of the disease. CONCLUSIONS: Liver cell transplantation can achieve donor cell engraftment in humans in a significant proportion, leading to sustained metabolic and clinical control with psychomotor catch-up.


Graft Survival , Hepatocytes/transplantation , Metabolism, Inborn Errors/surgery , Ammonia/blood , Argininosuccinate Lyase/metabolism , Argininosuccinic Acid/blood , Argininosuccinic Acid/urine , Argininosuccinic Aciduria , Child Development , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , Liver/physiopathology , Liver Function Tests , Male , Metabolism, Inborn Errors/physiopathology , Metabolism, Inborn Errors/psychology , Psychomotor Performance , Time Factors , Transplantation Chimera , alpha-Fetoproteins/metabolism
16.
Eur J Cancer ; 42(1): 65-72, 2006 Jan.
Article En | MEDLINE | ID: mdl-16343894

Conclusive identification of RB1 mutations in retinoblastoma is predicted to improve the clinical management of affected children and relatives. However, despite clear clinical benefits, RB1 screening remains difficult, most of the alterations being unique and randomly distributed throughout the entire coding sequence. In this report, we present the results of a constitutional RB1 analysis undertaken in our institution over the last four years. The detection of RB1 gene deletion or mutation was performed by Southern blot and sequence analyses in 73 patients (including three families with 2, 3 and 3 probands, respectively). Complementary constitutional chromosome and fluorescent in situ hybridization (FISH) analyses of RB1 gene were applied in cases where hereditary retinoblastoma was suspected despite negative detection. Altogether, germline abnormalities were found in 11% (4/36 patients) of sporadic unilateral retinoblastoma (median age, 21.5 months) and 86% (32/37 patients) of sporadic bilateral or positive familial history retinoblastoma (median age, 5 months). The spectrum of germline alterations found in 31 distinct families included 12 nonsense mutations (39%); 10 point insertions or deletions with frameshift (32%); 4 mutations and 1 deletion affecting splice sites (16%); 2 missense mutations (6%); and 2 large deletions (6%). A total of 15 mutations have not been previously reported. In this small series, splicing mutations were associated with bilateral disease whilst most of the frameshift mutations were identified in patients with an early age at diagnosis, bilateral disease or hereditary forms of the disease. This study confirms that screening for constitutional RB1 mutation should become an integral part of current management of any patient affected by retinoblastoma irrespective of the tumour laterality and familial background.


Genes, Retinoblastoma/genetics , Germ-Line Mutation/genetics , Retinal Neoplasms/genetics , Retinoblastoma Protein/genetics , Retinoblastoma/genetics , Adult , Age Factors , Blotting, Southern , Female , Humans , Male , Pedigree , Retinal Neoplasms/pathology , Retinoblastoma/pathology
17.
Exp Hematol ; 33(10): 1147-59, 2005 Oct.
Article En | MEDLINE | ID: mdl-16219537

OBJECTIVE: Determine the molecular defects underlying the CD3(-)CD4(+) T-cell phenotype and persistence of this clonal population in patients with hypereosinophilic syndrome. PATIENTS AND METHODS: Patients in this study suffer from the lymphocytic variant of hypereosinophilic syndrome distinguished by a CD3(-)CD4(+) T-cell clone that overexpresses Th2 cytokines upon activation and thereby provokes the eosinophilia. Interleukin-2-dependent CD3(-)CD4(+) T-cell lines were derived from patient blood at various disease stages and used to investigate the molecular modifications correlated with their abnormal phenotype. RESULTS: We demonstrate that the CD3(-)CD4(+) T cells, characterized by a clonal TCRbeta gene rearrangement, maintained the same immunophenotype over the 6-year period of our study, during which one patient progressed from premalignant disease to CD3(-)CD4(+) T-cell lymphoma. We show that a specific loss of CD3gamma gene transcripts is responsible for the defect in TCR/CD3 surface expression. In addition, the level of NFATc2 binding to NFAT motifs in the CD3gamma gene promoter was greatly increased in the abnormal T cells. Our studies indicate that CD3gamma promoter activity can be positively influenced by NFATc1 plus NF-kappaB p50 and negatively regulated by NFATc2 containing complexes. We show that in patients' CD3(-)CD4(+) T cells, an increase in nuclear NFATc2 occurs in parallel with a decrease in NFATc1 and NF-kappaB gene expression. CONCLUSION: Hypereosinophilic syndrome joins the growing number of pathological conditions where a defect in surface expression and/or function of the TCR/CD3 complex results from altered regulation of CD3gamma gene expression.


CD3 Complex/biosynthesis , Gene Expression Regulation , Hypereosinophilic Syndrome/metabolism , NFATC Transcription Factors/metabolism , Th2 Cells/metabolism , Transcription, Genetic , Adult , CD3 Complex/genetics , Cell Line , Cytokines/metabolism , Female , Gene Expression Regulation/genetics , Humans , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/pathology , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Response Elements/genetics , Th2 Cells/pathology , Transcription, Genetic/genetics
18.
Haematologica ; 90(6): 753-65, 2005 Jun.
Article En | MEDLINE | ID: mdl-15951288

BACKGROUND AND OBJECTIVES: The lymphocytic variant of hypereosinophilic syndrome (LV-HES) is an underrated disease defined by the monoclonal proliferation of interleukin-5 secreting T-cells. This disease is distinguished by a period of chronic lymphoproliferation without clinical transformation, which is frequently a precursor to T-cell lymphoma. In this study, LV-HES was used as a model of pre-malignancy to identify specific marker(s) predictive of the potential for malignant transformation. DESIGN AND METHODS: The karyotypic abnormalities detected in the abnormal CD3-CD4+ T cells were further characterized by fluorescent in situ hybridization. A multi-step retrospective analysis was performed on successive blood samples during a six-year follow up to correlate the evolution of cytogenetic changes with clinical progression. Expression array analysis was used to investigate the effect of these chromosomal aberrations on gene expression. RESULTS: A 6q deletion was detected in the two LV-HES patients during their chronic disease phase. An additional 10p deletion was found alone or in association with the 6q defect in one patient prior to the development of a CD3-CD4+ T-cell lymphoma six years after diagnosis. We show that the 6q but not the 10p deletion is both stable and persistent throughout the chronic disease, finally emerging as the predominant aberration in the lymphoma cells. Six genes mapped to the 6q-deleted region displayed altered gene expression profiles both in the chronic and malignant disease phases. INTERPRETATION AND CONCLUSIONS: Our data suggest that the 6q deletion represents an early cytogenetic marker for T-cell transformation.


CD3 Complex/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Chromosome Aberrations , Chromosomes, Human, Pair 6 , Hypereosinophilic Syndrome/genetics , Adult , Base Sequence , Chromosomes, Artificial, Bacterial , Chromosomes, Human, Pair 10 , Female , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , Lymphocytes/pathology , Molecular Sequence Data
19.
Blood ; 99(11): 4154-9, 2002 Jun 01.
Article En | MEDLINE | ID: mdl-12010820

CD4(+), CD56(+) DC2 malignancies constitute a novel disease entity, which has recently been shown to arise from a transformed lymphoid-related plasmacytoid dendritic cell (DC2). Diagnosis is primarily based on a particular immunophenotype with tumor cells expressing CD4 and CD56 antigens in the absence of common lymphoid or myeloid lineage markers. Little is currently known about the cytogenetic features of this disease entity. In this setting, the Groupe Français de Cytogénétique Hématologique (GFCH) initiated a cytogenetic study of 18 adults and 3 children with CD4(+), CD56(+) DC2 acute leukemia using conventional and fluorescence in situ hybridization/24-color karyotyping. Clonal, mostly complex chromosome aberrations were found in 14 patients (66%). Six major recurrent chromosomal targets were defined. These were 5q, 12p, 13q, 6q, 15q, and 9, which were involved in 72% (5q), 64% (12p and 13q), 50% (6q), 43% (15q), and 28% (monosomy 9) of cases, respectively. Cytogenetic features can be summarized as follows: (1) gross genomic imbalances (mostly losses) predominate, (2) no single anomaly can be considered as specific, whereas their combination/accumulation is, and (3) both lymphoid and myeloid lineage-associated rearrangements are observed in unusual combinations in the same cell. This is suggestive of complex multistep tumorigenic mechanisms and is supportive of the hypothesis that CD4(+), CD56(+) DC2 acute leukemia may arise from an undifferentiated nonmyeloid nonlymphoid progenitor cell. In conclusion, the present study documents for the first time the existence of a characteristic cytogenetic profile for this novel disease entity.


CD4 Antigens/analysis , CD56 Antigen/analysis , Chromosome Aberrations , Leukemia/genetics , Acute Disease , Adolescent , Aged , Aged, 80 and over , Child , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia/immunology , Male , Middle Aged
...