Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Acta Haematol ; 144(1): 58-65, 2021.
Article in English | MEDLINE | ID: mdl-32512574

ABSTRACT

INTRODUCTION: Multiple myeloma (MM) is uncommon in persons younger than 50 years of age. The presenting features in this age group are unclear. METHODS: We analyzed a cohort of patients <50 years of age with MM treated in our center. RESULTS: Twenty-three patients at a median age of 41.5 years (range 27-49) were analyzed. Patients presented at International Staging System (ISS) I-II (79%), had high frequency of bone lytic lesions (89%), extramedullary disease (26%), light-chain myeloma (45%), and translocation t(11;14) (68%). The subpopulation of patients carrying t(11;14) were younger (p = 0.025). This subgroup had higher bone marrow infiltration of plasma cells (75 vs. 47.5%), higher incidence of proteinuria (2.9 vs. 0.19 g/day), and poorer response to therapy: 85.7% of patients achieving complete/very good partial remission after induction therapy did not have t(11;14). A trend toward inferior progression-free survival (PFS) was observed in patients with t(11; 14) compared to patients without this translocation (median PFS 18 and 36 months, respectively). DISCUSSION/CONCLUSION: Translocation t(11; 14) seems to be more prevalent in young myeloma patients. Young myeloma patients and especially those who harbor translocation t(11; 14) may represent a distinct clinical entity that confers a poor outcome.


Subject(s)
Multiple Myeloma/epidemiology , Adult , Age Factors , Age of Onset , Biomarkers , Chromosome Aberrations , Combined Modality Therapy , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/etiology , Multiple Myeloma/therapy , Neoplasm Staging , Prognosis , Public Health Surveillance , Treatment Outcome , Young Adult
2.
Oncogene ; 39(16): 3396-3410, 2020 04.
Article in English | MEDLINE | ID: mdl-32103168

ABSTRACT

E3 ubiquitin ligases (E3s) play essential roles in the maintenance of tissue homeostasis under normal and stress conditions, as well as in disease states, particularly in cancer. However, the role of E3s in the initiation of human tumors is poorly understood. Previously, we reported that genetic ablation of the HECT-type E3 ubiquitin ligase Smurf2 induces carcinogenesis in mice; but whether and how these findings are pertinent to the inception of human cancer remain unknown. Here we show that SMURF2 is essential to protect human dermal fibroblasts (HDFs) from malignant transformation, and its depletion converts HDFs into tumorigenic entity. This phenomenon was associated with the radical changes in chromatin structural and epigenetic landscape, dysregulated gene expression and cell-cycle control, mesenchymal-to-epithelial transition and impaired DNA damage response. Furthermore, we show that SMURF2-mediated tumor suppression is interlinked with SMURF2's ability to regulate the expression of two central chromatin modifiers-an E3 ubiquitin ligase RNF20 and histone methyltransferase EZH2. Silencing these factors significantly reduced the growth and transformation capabilities of SMURF2-depleted cells. Finally, we demonstrate that SMURF2-compromised HDFs are highly tumorigenic in nude mice. These findings suggest the critical role that SMURF2 plays in preventing malignant alterations, chromosomal instability and cancer.


Subject(s)
Carcinogenesis/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Chromatin/genetics , Chromosomal Instability/genetics , Dermis/metabolism , Dermis/pathology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Silencing , Humans , Mice , Mice, Nude , Neoplasms/pathology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitination/genetics
3.
Acta Haematol ; 141(2): 119-127, 2019.
Article in English | MEDLINE | ID: mdl-30726835

ABSTRACT

BACKGROUND: The WHO defined myeloid and lymphoid neoplasms (MLN) with eosinophilia associated with PDGFRB, PDGFRA, FGFR1 rearrangements as a new entity in 2016. PDGFRB-rearranged MLN sensitive to imatinib were described in adult patients. We report the first pediatric patient with PDGFRB-rearranged myeloproliferative disorder associated with T-lymphoblastic lymphoma bearing the t(5; 14)(q33;q32) translocation who was successfully treated with imatinib only. Methods/Aims: Analysis of bone marrow and peripheral blood cells by fluorescent in situ hybridization identified the PDGFRB partner as CCDC88C. Whole genome sequencing of the patient's DNA identified the exact junction site, confirmed by PCR amplification and Sanger sequencing. A real-time quantitative PCR assay was designed to quantify the fused CCDC88C-PDGFRB product. RESULTS: A 2.5-year-old boy was diagnosed with myeloproliferative disorder and eosinophilia associated with lymphoblastic lymphoma both bearing the CCDC88C-PDGFRB fusion. Imatinib therapy resulted in rapid clinical, hematological, and cytogenetic response. Molecular response to treatment was monitored by a real-time PCR assay specific for the CCDC88C- PDGFRB fusion. CONCLUSION: This is the first description of MLN with eosinophilia in the pediatric age group. Response to treatment with imatinib only was monitored by specific quantitative PCR assay with sustained remission lasting 5.5 years from diagnosis.


Subject(s)
Imatinib Mesylate/therapeutic use , Intracellular Signaling Peptides and Proteins/genetics , Microfilament Proteins/genetics , Myeloproliferative Disorders/drug therapy , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/genetics , Base Sequence , Child, Preschool , Humans , In Situ Hybridization, Fluorescence , Karyotype , Male , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Whole Genome Sequencing
4.
Genes Chromosomes Cancer ; 58(8): 516-520, 2019 08.
Article in English | MEDLINE | ID: mdl-30675954

ABSTRACT

The current study evaluated the prognostic significance of the monoallelic deletion of the whole locus of the immunoglobulin heavy-chain (w_del(IGH)) gene compared to translocations t(4;14) and t(14;16) among newly diagnosed multiple myeloma (MM) patients. We retrospectively analyzed clinical (age, gender, and staging) and laboratory data at diagnosis and the overall survival (OS) of 255 newly diagnosed MM patients carrying w_del(IGH) or translocations t(4;14) or t(14;16). Bone marrow samples were examined by morphological and sequential interphase fluorescense in situ hybridization analyses. Among 255 patients, 117 (45.8%) had w_del(IGH), 99 (38.8%) had t(4;14), and 39 (15.3%) had t(14;16). Mean age was 61.6 ± 11.6 years. Groups did not differ significantly in age, gender, or lactate dehydrogenase levels. Patients in the w_del(IGH) group presented more frequently at International Staging System stage I than at stage II/III. Patients in the w_del(IGH) group had significantly fewer additional chromosomal aberrations (1.58) than the other two groups (2.3 and 2.13 in the del(IGH), t(14;16) and t(4;14) groups, respectively, P < 0.0001). Furthermore, the w_del(IGH) group had significantly longer estimated median OS (9.47 years) compared to those with translocations t(14;16) (3.02 years, P = 0.002) or t(4;14) (4.18 years, P = 0.001), respectively. These findings suggest a potential prognostic significance of monoallelic deletion of IGH among these patients. Additional studies are needed to better understand the nature and mechanism of this prognostic factor.


Subject(s)
Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 4 , Gene Deletion , Genes, Immunoglobulin Heavy Chain , Multiple Myeloma/mortality , Translocation, Genetic , Aged , Female , Follow-Up Studies , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Neoplasm Staging , Odds Ratio , Prognosis , Survival Analysis
5.
Leuk Lymphoma ; 60(3): 764-771, 2019 03.
Article in English | MEDLINE | ID: mdl-30187812

ABSTRACT

Chromosome banding analysis (CBA) in myelodysplastic syndromes (MDS) remains the 'gold standard' for identification of chromosomal abnormalities, while interphase fluorescence in-situ hybridization (I-FISH) is mainly used to complement CBA. This study, retrospectively, evaluated CBA and I-FISH results in 600 patients with suspected MDS and determined the effect of CBA/FISH reallocation on IPSS-R. Our result demonstrated that in 7/586 (1.2%) patients with satisfactory karyotype, I-FISH provided additional information. In 25/453 (5.5%) of the patients with normal I-FISH, CBA detected chromosomal abnormalities, and in 68/147 (46%) of the patients with abnormal I-FISH, CBA detected additional chromosomal aberrations. When 5q- aberration was alone or accompanied by additional abnormalities by I-FISH, CBA revealed a complex karyotype (16/25;64%, 35/43;81%, respectively). Our results suggest that in cases of karyotype failure, if I-FISH is used alone, patients are at risk of being misclassified into the wrong cytogenetic risk groups and a repeat sample for CBA should be attempted.


Subject(s)
Cytogenetics , In Situ Hybridization, Fluorescence , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Biopsy , Bone Marrow/pathology , Chromosome Aberrations , Chromosome Banding , Cytogenetics/methods , Cytogenetics/standards , Female , Humans , In Situ Hybridization, Fluorescence/methods , In Situ Hybridization, Fluorescence/standards , Karyotyping , Male , Prognosis , Reproducibility of Results , Sensitivity and Specificity
7.
Oncotarget ; 8(16): 27380-27392, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28423708

ABSTRACT

Advanced ovarian cancer is an incurable disease. Thus, novel therapies are required. We wished to identify new therapeutic targets for ovarian cancer. ShRNA screen performed in 42 ovarian cancer cell lines identified the centriolar replication factor STIL as an essential gene for ovarian cancer cells. This was verified in-vivo in orthotopic human ovarian cancer mouse models. STIL depletion by administration of siRNA in neutral liposomes resulted in robust anti-tumor effect that was further enhanced in combination with cisplatin. Consistent with this finding, STIL depletion enhanced the extent of DNA double strand breaks caused by DNA damaging agents. This was associated with centrosomal depletion, ongoing genomic instability and enhanced formation of micronuclei. Interestingly, the ongoing DNA damage was not associated with reduced DNA repair. Indeed, we observed that depletion of STIL enhanced canonical homologous recombination repair and increased BRCA1 and RAD51 foci in response to DNA double strand breaks. Thus, inhibition of STIL significantly enhances the efficacy of DNA damaging chemotherapeutic drugs in treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA Breaks, Double-Stranded , DNA Repair , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Targeted Therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA Interference , RNA, Small Interfering/genetics , Recombinational DNA Repair , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
8.
Int J Cancer ; 140(4): 864-876, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27813122

ABSTRACT

Quercetin (Que) is an abundant flavonoid in the human diet and high-concentration food supplement with reported pro- and anti-carcinogenic activities. Topoisomerase II (TopoII) inhibition and subsequent DNA damage induction by Que was implicated in the mixed lineage leukemia gene (MLL) rearrangements that can induce infant and adult leukemias. This notion raised concerns regarding possible genotoxicities of Que in hematopoietic stem and progenitor cells (HSPCs). However, molecular targets mediating Que effects on DNA repair relevant to MLL translocations have not been defined. In this study we describe novel and potentially genotoxic Que activities in suppressing non-homologous end joining and homologous recombination pathways downstream of MLL cleavage. Using pharmacological dissection of DNA-PK, ATM and PI3K signalling we defined PI3K inhibition by Que with a concomitant decrease in the abundance of key DNA repair genes to be responsible for DNA repair inhibition. Evidence for the downstream TopoII-independent mutagenic potential of Que was obtained by documenting further increased frequencies of MLL rearrangements in human HSPCs concomitantly treated with Etoposide and Que versus single treatments. Importantly, by engaging a tissue engineered placental barrier, we have established the extent of Que transplacental transfer and hence provided the evidence for Que reaching fetal HSPCs. Thus, Que exhibits genotoxic effects in human HSPCs via different mechanisms when applied continuously and at high concentrations. In light of the demonstrated Que transfer to the fetal compartment our findings are key to understanding the mechanisms underlying infant leukemia and provide molecular markers for the development of safety values.


Subject(s)
Cell Transformation, Neoplastic/drug effects , DNA Damage , DNA Repair/drug effects , DNA Topoisomerases, Type II/physiology , Hematopoietic Stem Cells/drug effects , Histone-Lysine N-Methyltransferase/genetics , Leukemia/chemically induced , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphoinositide-3 Kinase Inhibitors , Quercetin/toxicity , Signal Transduction/drug effects , Topoisomerase II Inhibitors/toxicity , Adult , Ascorbic Acid/pharmacology , Cell Culture Techniques , Cells, Cultured , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Etoposide/pharmacology , Female , Genistein/pharmacology , Histones/analysis , Humans , Infant , Leukemia/genetics , Maternal-Fetal Exchange , Phosphatidylinositol 3-Kinases/physiology , Pregnancy
9.
Cytometry A ; 89(7): 644-52, 2016 07.
Article in English | MEDLINE | ID: mdl-27315046

ABSTRACT

B-cell chronic lymphocytic leukaemia (B-CLL) and B-cell precursor acute lymphoblastic leukaemia (B-ALL) are the most common type of leukaemia in adults and children, respectively. Today, fluorescence in situ hybridization (FISH) is the standard for detecting chromosomal aberrations that reflect adverse and favorable outcome. This study revealed a new, simple, and fast diagnostic tool to detect pathological cells by measuring and imaging the fluorescence lifetime (FLT) using FLT imaging microscopy (FLIM) of the peripheral blood (PB) cells of B-CLL samples that were labeled with the DNA binder, DAPI. The FLT of DAPI in healthy individuals was found to be 2.66 ± 0.12 ns. In contrast, PB cells of B-CLL and BM cells of B-ALL patients were characterized by a specific group distribution of the FLT values. The FLT of DAPI was divided into four subgroups, relative to 2.66 ns: short+, normal, prolonged, and prolonged+. These alterations could be related to different chromatin arrangements of B-CLL and B-ALL interphase nuclei. Notably, extremely long FLT of nuclear DAPI correlate with the presence of extra chromosome 12, while moderate increases compared to normal characterize the deletion of p53. Such correlations potentially enable a FLT-based rapid automatic diagnosis and classification of B-CLL even when the frequency of genetic and chromosomal abnormalities is low. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/diagnostic imaging , Optical Imaging/methods , Cell Nucleus/pathology , Fluorescent Dyes , Humans , Indoles
10.
Genes Chromosomes Cancer ; 55(9): 710-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27152944

ABSTRACT

The most common translocation in multiple myeloma (MM) is t(11;14)(q13;q32). According to several studies, this translocation represents a unique subset of patients with relatively favorable outcomes. Using combined analyses of morphology and fluorescence in situ hybridization (I-FISH), we examined the co-occurrence rates of t(11;14) with seven chromosomal aberrations (CAs), del(13q), del(17p), del(1p), gain(1q), multiple gains(1q), del(16q), and del(IGH), and assessed the effect of the different combinations on patient outcomes, with overall survival (OS) as the main outcome measure. Bone marrow samples and clinical data from 212 patients with MM with t(11;14) were analyzed. At least two additional CAs were found in 35% (75/205) of patients and a strong correlation between specific CAs. The occurrence of three CAs [multiple gains of (1q) (HR = 6.94, P = 0.001), del(1p) (HR = 4.47, P = 0.008), and del(IGH) (HR = 2.38, P = 0.002)] exerted a profoundly deleterious effect on median OS when compared with patients with t(11;14) only. Del(17p) and del(13q) have also exerted a deleterious effect albeit to a lesser extent (HR = 2.05, P = 0.07 and HR = 1.81, P = 0.03, respectively). When compared with t(11;14) alone, the addition of certain CAs lead to worse outcomes. These findings may have important clinical and biological implications. Patients with coexisting adverse lesions and t(11;14) may be considered at high risk and managed accordingly. © 2016 Wiley Periodicals, Inc.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Translocation, Genetic/genetics , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Female , Follow-Up Studies , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate
11.
Head Neck ; 38 Suppl 1: E741-6, 2016 04.
Article in English | MEDLINE | ID: mdl-25900614

ABSTRACT

BACKGROUND: Oral lichen planus (OLP) carries an increased risk for malignant transformation with aneuploid cells (ACs) being found in brush samples of a quarter of patients with OLP. METHODS: Patients with OLP were followed and repeated brush samples were simultaneously analyzed for morphology and fluorescent in situ hybridization (FISH) using centromeric probes for chromosomes 2 and 8. RESULTS: Three patients with a high proportion of ACs developed oral cancer. Fifteen patients had ≥1% ACs (13 in affected sites and 2 in nonaffected sites), whereas only 2 of the 15 patients with <1% ACs in the first sample had ≥1% ACs in the second sample. A strong positive correlation between the results of the initial and repeated samples was found. CONCLUSION: High proportion of ACs in brush samples from patients with OLP may imply an impending malignant transformation. As FISH analysis is consistent over time, it can be used to identify a subgroup of patients who would require close follow-up. © 2015 Wiley Periodicals, Inc. Head Neck 38: E741-E746, 2016.


Subject(s)
Aneuploidy , Lichen Planus, Oral/genetics , Mouth Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 8/genetics , Female , Follow-Up Studies , Humans , In Situ Hybridization, Fluorescence , Lichen Planus, Oral/pathology , Male , Middle Aged , Mouth Neoplasms/genetics
13.
Mol Cytogenet ; 7(1): 9, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24472424

ABSTRACT

BACKGROUND: Polyploidy has been recognized for many years as an important hallmark of cancer cells. Polyploid cells can arise through cell fusion, endoreplication and abortive cell cycle. The inner nuclear membrane protein LAP2ß plays key roles in nuclear envelope breakdown and reassembly during mitosis, initiation of replication and transcriptional repression. Here we studied the function of LAP2ß in the maintenance of cell ploidy state, a role which has not yet been assigned to this protein. RESULTS: By knocking down the expression of LAP2ß, using both viral and non-viral RNAi approaches in osteosarcoma derived U2OS cells, we detected enlarged nuclear size, nearly doubling of DNA content and chromosomal duplications, as analyzed by fluorescent in situ hybridization and spectral karyotyping methodologies. Spectral karyotyping analyses revealed that near-hexaploid karyotypes of LAP2ß knocked down cells consisted of not only seven duplicated chromosomal markers, as could be anticipated by genome duplication mechanism, but also of four single chromosomal markers. Furthermore, spectral karyotyping analysis revealed that both of two near-triploid U2OS sub-clones contained the seven markers that were duplicated in LAP2ß knocked down cells, whereas the four single chromosomal markers were detected only in one of them. Gene expression profiling of LAP2ß knocked down cells revealed that up to a third of the genes exhibiting significant changes in their expression are involved in cancer progression. CONCLUSIONS: Our results suggest that nuclear fusion mechanism underlies the polyploidization induction upon LAP2ß reduced expression. Our study implies on a novel role of LAP2ß in the maintenance of cell ploidy status. LAP2ß depleted U2OS cells can serve as a model to investigate polyploidy and aneuploidy formation by nuclear fusion mechanism and its involvement in cancerogenesis.

14.
J Pediatr Hematol Oncol ; 35(4): 303-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23612381

ABSTRACT

The differential diagnosis of hypereosinophilia includes both primary (clonal and idiopathic) and secondary medical conditions. Here we raise the awareness of physicians to the unusual causes of hypereosinophilic states and describe the molecular assays used in the diagnosis of hypereosinophilia. Two unusual cases of hypereosinophilia in children that were initially misdiagnosed are reported. T-cell receptor gene rearrangement, skewed X inactivation, fluorescence in situ hybridization analysis, and chromosomal karyotyping were used to reach the final correct diagnosis. Both patients displayed significant eosinophilia and were initially misdiagnosed as having parasitic infection. Nonspecific T-cell clonal expansion was diagnosed in 1 patient based on the clonality of the T-cell receptor variable γ-chain and the skewed chromosome inactivation. The second patient was diagnosed with B-lineage acute lymphoblastic leukemia with a translocation (5;14) (q13;q32) that is well known to be associated with hypereosinophilia. The level of awareness to clonal expansion of WBC subsets which can cause hypereosinophilia should be high when evaluating a patient with extreme eosinophilia. Advanced molecular assays to detect clonal expansion should be used to exclude aberrant clonal processes in such patients.


Subject(s)
Eosinophilia/diagnosis , Child , Child, Preschool , Diagnosis, Differential , Eosinophilia/genetics , Female , Gene Order , Humans , In Situ Hybridization, Fluorescence , Male , Receptors, Antigen, T-Cell/genetics
15.
Clin Dev Immunol ; 2012: 261470, 2012.
Article in English | MEDLINE | ID: mdl-23243423

ABSTRACT

INTRODUCTION: Patients with severe combined immunodeficiency (SCID) may present with residual circulating T cells. While all cells are functionally deficient, resulting in high susceptibility to infections, only some of these cells are causing autoimmune symptoms. METHODS: Here we compared T-cell functions including the number of circulating CD3(+) T cells, in vitro responses to mitogens, T-cell receptor (TCR) repertoire, TCR excision circles (TREC) levels, and regulatory T cells (Tregs) enumeration in several immunodeficinecy subtypes, clinically presenting with nonreactive residual cells (MHC-II deficiency) or reactive cells. The latter includes patients with autoreactive clonal expanded T cell and patients with alloreactive transplacentally maternal T cells. RESULTS: MHC-II deficient patients had slightly reduced T-cell function, normal TRECs, TCR repertoires, and normal Tregs enumeration. In contrast, patients with reactive T cells exhibited poor T-cell differentiation and activity. While the autoreactive cells displayed significantly reduced Tregs numbers, the alloreactive transplacentally acquired maternal lymphocytes had high functional Tregs. CONCLUSION: SCID patients presenting with circulating T cells show different patterns of T-cell activity and regulatory T cells enumeration that dictates the immunodeficient and autoimmune manifestations. We suggest that a high-tolerance capacity of the alloreactive transplacentally acquired maternal lymphocytes represents a toleration advantage, yet still associated with severe immunodeficiency.


Subject(s)
Severe Combined Immunodeficiency/immunology , T-Lymphocytes, Regulatory/immunology , Cell Differentiation/immunology , Humans , Immune Tolerance/immunology , Infant , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology
16.
Exp Hematol Oncol ; 1(1): 24, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-23211092

ABSTRACT

Extreme swing of phosphor from severe hyperphosphatemia to severe hypophosphatemia in a patient with blast crisis of myeloid origin was the result of imbalance between massive apoptosis of leukemic cells in the context of spontaneous tumor lysis syndrome and massive production of leukemic cells with only 1% of blast in peripheral blood. The mutated p53 protein suggested acting as oncogene in the presented case and possibly affecting phosphor status.

17.
Cancer Res ; 72(24): 6403-13, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23047867

ABSTRACT

Mesenchymal stromal cells (MSC) are used extensively in clinical trials; however, the possibility that MSCs have a potential for malignant transformation was raised. We examined the genomic stability versus the tumor-forming capacity of multiple mouse MSCs. Murine MSCs have been shown to be less stable and more prone to malignant transformation than their human counterparts. A large series of independently isolated MSC populations exhibited low tumorigenic potential under syngeneic conditions, which increased in immunocompromised animals. Unexpectedly, higher ploidy correlated with reduced tumor-forming capacity. Furthermore, in both cultured MSCs and primary hepatocytes, polyploidization was associated with a dramatic decrease in the expression of the long noncoding RNA H19. Direct knockdown of H19 expression in diploid cells resulted in acquisition of polyploid cell traits. Moreover, artificial tetraploidization of diploid cancer cells led to a reduction of H19 levels, as well as to an attenuation of the tumorigenic potential. Polyploidy might therefore serve as a protective mechanism aimed at reducing malignant transformation through the involvement of the H19 regulatory long noncoding RNA.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Silencing/physiology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Polyploidy , RNA, Long Noncoding/genetics , Animals , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Genomic Instability , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms/genetics , RNA, Long Noncoding/antagonists & inhibitors
18.
Cytokine ; 57(2): 290-3, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22169779

ABSTRACT

Imatinib (IM) is the current first line treatment for chronic myeloid leukemia (CML). However, the disease will progress in the majority of patients pausing IM. IFN-α may intensify the response and increase the percentage of patients maintaining remission after IM cessation. Eleven patients with stable (≥ 2 years) complete cytogenetic responses (CCyR) on IM therapy were recruited to the study. They were administered Peg-IFN-α for 9 months before and for 3 months following IM discontinuation. During the 12 months of Peg-IFN-α therapy the remission status improved in five (45%) of the patients. Six (55%) of the patients experienced cytogenetic relapses at a median period of 8 months (range 2-33) after IM withdrawal. All six patients regained CCyR following IM restart. With a median follow up of 47 months (range 35-50), five (45%) out of the 11 studied patients maintain cytogenetic response off IM therapy. The role of Peg-IFN-α in patients pausing IM is to be further evaluated.


Subject(s)
Antineoplastic Agents/therapeutic use , Antiviral Agents/therapeutic use , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Withholding Treatment , Adult , Benzamides , Demography , Female , Humans , Imatinib Mesylate , Male , Middle Aged , Treatment Outcome
19.
Genome Biol ; 12(6): R61, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21714908

ABSTRACT

BACKGROUND: Chromosomal aneuploidy, that is to say the gain or loss of chromosomes, is the most common abnormality in cancer. While certain aberrations, most commonly translocations, are known to be strongly associated with specific cancers and contribute to their formation, most aberrations appear to be non-specific and arbitrary, and do not have a clear effect. The understanding of chromosomal aneuploidy and its role in tumorigenesis is a fundamental open problem in cancer biology. RESULTS: We report on a systematic study of the characteristics of chromosomal aberrations in cancers, using over 15,000 karyotypes and 62 cancer classes in the Mitelman Database. Remarkably, we discovered a very high co-occurrence rate of chromosome gains with other chromosome gains, and of losses with losses. Gains and losses rarely show significant co-occurrence. This finding was consistent across cancer classes and was confirmed on an independent comparative genomic hybridization dataset of cancer samples. The results of our analysis are available for further investigation via an accompanying website. CONCLUSIONS: The broad generality and the intricate characteristics of the dichotomy of aneuploidy, ranging across numerous tumor classes, are revealed here rigorously for the first time using statistical analyses of large-scale datasets. Our finding suggests that aneuploid cancer cells may use extra chromosome gain or loss events to restore a balance in their altered protein ratios, needed for maintaining their cellular fitness.


Subject(s)
Aneuploidy , Chromosome Aberrations , Karyotype , Neoplasms/genetics , Cluster Analysis , Data Mining , Humans , Internet , Neoplasms/classification , User-Computer Interface
20.
Cancer Cytopathol ; 119(4): 235-46, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21557514

ABSTRACT

BACKGROUND: Most cases of laryngeal cancer are preceded by precursor lesions which, if left untreated, can progress toward an invasive cancer. The objective of this study was to investigate the presence of chromosomal numerical aberrations in cells that were collected by noninvasive brush sampling from laryngeal lesions. METHODS: Laryngeal brush samples from 52 patients were analyzed simultaneously for morphology and fluorescence in situ hybridization (FISH) using centromeric probes for chromosome 17, chromosome 8, and a locus-specific instability (LSI) v-myc avian myelocytomatosis viral oncogene homolog (myc) proto-oncogene protein (C-MYC) probe for the MYC gene. The patients were divided according to histopathologic diagnosis. Group 1 included patients with squamous cell carcinoma, carcinoma in situ, and severe dysplasia; Group 2 included patients with moderate dysplasia, mild dysplasia, and hyperplasia; and Group 3 included patients with benign nondysplastic lesions. RESULTS: The proportion of cells with MYC and chromosome 8 gains demonstrated significant trends toward being the highest in Group 1 and the lowest in Group 3 (P = .001 and P = .003, respectively). No significant trend was observed for chromosome 17. Mann-Whitney Bonferroni-corrected analyses revealed that the most significant contribution was the difference between Groups 1 and 3 (P = .0195 for MYC gains and P = .036 for chromosome 8 gains). When using a cutoff point of 4% aneuploid cells (ACs), both MYC and chromosome 8 differed significantly between groups (P = .030 and P = .037, respectively). CONCLUSIONS: The current results suggested that FISH analysis of brush samples obtained noninvasively from suspicious laryngeal lesions can augment the clinical examination in predicting the nature of the lesions and can aid clinicians in monitoring and follow-up of high-risk patients. Cancer (Cancer Cytopathol) 2011. © 2011 American Cancer Society.


Subject(s)
Aneuploidy , Carcinoma in Situ/diagnosis , Hyperplasia/diagnosis , Laryngeal Neoplasms/diagnosis , Larynx/pathology , Precancerous Conditions/diagnosis , Adult , Aged , Carcinoma in Situ/genetics , Case-Control Studies , Female , Follow-Up Studies , Humans , Hyperplasia/genetics , In Situ Hybridization, Fluorescence , Laryngeal Neoplasms/genetics , Male , Middle Aged , Precancerous Conditions/genetics , Prognosis , Proto-Oncogene Mas
SELECTION OF CITATIONS
SEARCH DETAIL
...