cyclin-d1 has been researched along with Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma* in 27 studies
1 review(s) available for cyclin-d1 and Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma
Article | Year |
---|---|
Therapy-related B lymphoblastic leukemia with t(4;11)(q21;q23)/AF4-MLL in a patient with mantle cell lymphoma after recent aggressive chemotherapy: a unique case report.
Mantle cell lymphoma (MCL) is a mature B-cell lymphoma associated with the hallmark translocation t(11;14)(q13;32), which involves the cyclin D1 (CCND1) and immunoglobin heavy chain (IgH) genes. It may transform to a more aggressive blastoid or pleomorphic variant, with or without acquisition of chromosomal abnormalities. MCL could also present with a leukemic phase with marked lymphocytosis. A literature search did not reveal any prior reports of MCL transforming to or followed by a B-cell lymphoblastic leukemia (B-ALL). Topics: Chromosomes, Human, Pair 11; Chromosomes, Human, Pair 4; Cyclin D1; DNA-Binding Proteins; Drug Therapy; Drug-Related Side Effects and Adverse Reactions; Gene Rearrangement; Histone-Lysine N-Methyltransferase; Humans; Immunoglobulin Heavy Chains; Lymphoma, Mantle-Cell; Male; Middle Aged; Myeloid-Lymphoid Leukemia Protein; Nuclear Proteins; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Transcriptional Elongation Factors; Translocation, Genetic; Treatment Outcome | 2014 |
1 trial(s) available for cyclin-d1 and Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma
Article | Year |
---|---|
Cyclin D1 expression in acute leukemia.
Disorders of the cell cycle regulatory machinery play a key role in the pathogenesis of cancer. Over expression of cyclin D1 protein has been reported in several solid tumors and certain lymphoid malignancies, but little is known about the involvement of cyclin D1 in acute leukemia.. In this study, we analyzed the expression of cyclin D1 at protein level in, 40 newly diagnosed patients with acute myeloid leukemia (AML), 10 patients with acute lymphoblastic leukemia (ALL), and 11 normal controls using flow cytometry.. The expression of cyclin D1 was not significantly different in AML group as compared to normal controls. On the other hand, over expression of cyclin D1 was evident in ALL group (4/10) as compared to that in healthy control. The ALL cases with cyclin D1 over expression were significantly correlated to blast cell counts in the peripheral blood and bone marrow (BM) but not with hemoglobin level, WBC, and platelets count. The ALL group with lymphadenopathy and organomegaly express significantly higher cyclin D1 over expression as compared to those without.. The biological value of cyclin D1 over expression might be different in AML and ALL. Topics: Adult; Biomarkers, Tumor; Bone Marrow; Cyclin D1; Female; Gene Expression Regulation, Leukemic; Humans; Leukemia, Myeloid, Acute; Male; Middle Aged; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2006 |
25 other study(ies) available for cyclin-d1 and Precursor-Cell-Lymphoblastic-Leukemia-Lymphoma
Article | Year |
---|---|
Significant Association of
This study investigated whether genetic variations in cyclin D1 (CCND1) are associated with susceptibility to childhood acute lymphoblastic leukemia (ALL).. A total of 266 childhood ALL cases and 266 healthy controls were genotyped for CCND1 rs9344 and rs678653.. There was a significant difference in the genotypic distribution of rs9344 between childhood ALL patients and healthy controls (p=0.0077). Compared to the AA genotype, AG and GG genotypes were associated with significantly decreased risks of childhood ALL with odds ratio (OR) of 0.65 [95% confidence interval (CI)=0.44-0.94, p=0.0234] and 0.45 (95%CI=0.26-0.78, p=0.0040), respectively. Supporting this, allelic frequency distributions between childhood ALL patients and controls was significantly different (OR=0.68, 95%CI=0.53-0.88, p=0.0025). There was no significant difference in the genotypic and allelic distributions of rs678653 between cases and controls.. CCND1 rs9344, but not rs678653, may serve as a predictive marker of susceptibility for childhood ALL. Topics: Alleles; Child; Child, Preschool; Cyclin D1; Female; Gene Frequency; Genetic Association Studies; Genetic Predisposition to Disease; Genotype; Humans; Male; Odds Ratio; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2021 |
B-Lymphoblastic Leukemia With Aberrant CD5 Expression.
B-acute lymphoblastic leukemia (B-ALL) is a neoplasm of precursor lymphoid cells committed to the B-lineage. Expression of CD5 is rare in B-ALL.. We studied the clinicopathologic, immunophenotypic, and molecular genetic features of 10 cases of B-ALL with aberrant CD5 expression, and compared with CD5-B-ALL.. B-ALL with aberrant CD5 expression is rare and predominantly affects men. Patients with CD5+ B-ALL had shorter median overall survival (21 vs 45 months, P = .0003). Expression of CD5 imposed a challenge in the differential diagnoses between B-ALL and other CD5+ B-cell lymphomas with blastic morphology. Dim CD20 and CD45, lack of surface immunoglobulin, expression of CD34 and TdT, negative immunostain for cyclin D1, and absence of t(11;14)(q13;q32) support a diagnosis of B-ALL.. CD5 expression is rare in B-ALL and associated with poor clinical outcome. CD5+ B-ALL represents a distinct entity that needs to be considered in the differential diagnoses of CD5+ B-cell lymphoproliferative disorders. Topics: Adolescent; Adult; Aged; B-Lymphocytes; CD5 Antigens; Child; Cyclin D1; Diagnosis, Differential; Female; Humans; Immunophenotyping; Male; Middle Aged; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Young Adult | 2021 |
Analytical evaluation of the clonoSEQ Assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma.
The clonoSEQ® Assay (Adaptive Biotechnologies Corporation, Seattle, USA) identifies and tracks unique disease-associated immunoglobulin (Ig) sequences by next-generation sequencing of IgH, IgK, and IgL rearrangements and IgH-BCL1/2 translocations in malignant B cells. Here, we describe studies to validate the analytical performance of the assay using patient samples and cell lines.. Sensitivity and specificity were established by defining the limit of detection (LoD), limit of quantitation (LoQ) and limit of blank (LoB) in genomic DNA (gDNA) from 66 patients with multiple myeloma (MM), acute lymphoblastic leukemia (ALL), or chronic lymphocytic leukemia (CLL), and three cell lines. Healthy donor gDNA was used as a diluent to contrive samples with specific DNA masses and malignant-cell frequencies. Precision was validated using a range of samples contrived from patient gDNA, healthy donor gDNA, and 9 cell lines to generate measurable residual disease (MRD) frequencies spanning clinically relevant thresholds. Linearity was determined using samples contrived from cell line gDNA spiked into healthy gDNA to generate 11 MRD frequencies for each DNA input, then confirmed using clinical samples. Quantitation accuracy was assessed by (1) comparing clonoSEQ and multiparametric flow cytometry (mpFC) measurements of ALL and MM cell lines diluted in healthy mononuclear cells, and (2) analyzing precision study data for bias between clonoSEQ MRD results in diluted gDNA and those expected from mpFC based on original, undiluted samples. Repeatability of nucleotide base calls was assessed via the assay's ability to recover malignant clonotype sequences across several replicates, process features, and MRD levels.. LoD and LoQ were estimated at 1.903 cells and 2.390 malignant cells, respectively. LoB was zero in healthy donor gDNA. Precision ranged from 18% CV (coefficient of variation) at higher DNA inputs to 68% CV near the LoD. Variance component analysis showed MRD results were robust, with expected laboratory process variations contributing ≤3% CV. Linearity and accuracy were demonstrated for each disease across orders of magnitude of clonal frequencies. Nucleotide sequence error rates were extremely low.. These studies validate the analytical performance of the clonoSEQ Assay and demonstrate its potential as a highly sensitive diagnostic tool for selected lymphoid malignancies. Topics: Bone Marrow; Cyclin D1; Gene Rearrangement; High-Throughput Nucleotide Sequencing; Humans; Immunoglobulin Heavy Chains; Immunoglobulin lambda-Chains; Immunoglobulins; Leukemia, Lymphocytic, Chronic, B-Cell; Limit of Detection; Multiple Myeloma; Neoplasm, Residual; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proto-Oncogene Proteins c-bcl-2; Reagent Kits, Diagnostic; Translocation, Genetic | 2020 |
Cyclin D1 G870A Polymorphism: Relation to the Risk of ALL Development, Prognosis Impact, and Methotrexate Cytotoxicity.
Cyclin D1 (CCND1) regulates cell cycle progression during the late G1 and S phase and takes part in methotrexate metabolism. It was hypothesized that CCND1 gene polymorphism affects acute lymphoblastic leukemia (ALL) development, prognosis and may relate to methotrexate cytotoxicity.. This study included 50 ALL patients and 50 healthy controls, CCND1 G870A polymorphism was studied in all items using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and evaluated methotrexate cytotoxicity for ALL patients using liver function tests before and after methotrexate treatment. We followed up patients for one year to determine disease-free survival (DFS) and overall survival (OS) and its relation to the CCND1 genotype.. We found that AA genotype and A allele have a higher risk of developing ALL compared to the control group. Additionally, we found no notable association between CCND1 variant and methotrexate cytotoxicity and no role of CCND1 polymorphism in ALL prognosis.. Our results suggested that CCND1 G870A polymorphism is associated with a high risk of ALL development. However, it has no role in ALL prognosis or methotrexate cytotoxicity. Topics: Adolescent; Antimetabolites, Antineoplastic; Case-Control Studies; Child; Child, Preschool; Cyclin D1; Drug-Related Side Effects and Adverse Reactions; Female; Follow-Up Studies; Genetic Predisposition to Disease; Humans; Infant; Liver Function Tests; Male; Methotrexate; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Survival Rate | 2020 |
Diagnostic utility of cyclin D1 in the diagnosis of small round blue cell tumors in children and adolescents.
Small round blue cell tumors (SRBCTs) of children and adolescents are often diagnostically challenging lesions. With the increasing diagnostic approach based on small biopsies, there is the need of specific immunomarkers that can help in the differential diagnosis among the different tumor histotypes to assure the patient a correct diagnosis for proper treatment. Based on our recent studies showing cyclin D1 overexpression in both Ewing sarcoma/primitive peripheral neuroectodermal tumor (EWS/pPNET) and peripheral neuroblastic tumors (neuroblastoma and ganglioneuroblastoma), we immunohistochemically assessed cyclin D1 immunoreactivity in 128 cases of SRBCTs in children and adolescents to establish its potential utility in the differential diagnosis. All cases of EWS/pPNET and the undifferentiated/poorly differentiated neuroblastomatous component of all peripheral neuroblastic tumors exhibited strong and diffuse nuclear staining (>50% of neoplastic cells) for cyclin D1. In contrast, this marker was absent from rhabdomyosarcoma (regardless of subtype) and lymphoblastic lymphoma (either B- or T-cell precursors), whereas it was only focally detected (<5% of neoplastic cells) in some cases of Wilms tumor (blastemal component) and desmoplastic small round cell tumor. Our findings suggest that cyclin D1 can be exploitable as a diagnostic adjunct to conventional markers in confirming the diagnosis of EWS/pPNET or neuroblastoma/ganglioneuroblastoma. Its use in routine practice may also be helpful for those cases of SRBCT with undifferentiated morphology that are difficult to diagnose after application of the conventional markers. Topics: Adolescent; Biomarkers, Tumor; Biopsy; Bone Neoplasms; Cell Differentiation; Child; Child, Preschool; Cyclin D1; Desmoplastic Small Round Cell Tumor; Diagnosis, Differential; Female; Ganglioneuroblastoma; Humans; Immunohistochemistry; Infant; Kidney Neoplasms; Male; Neuroblastoma; Neuroectodermal Tumors, Primitive, Peripheral; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Predictive Value of Tests; Retrospective Studies; Rhabdomyosarcoma; Sarcoma, Ewing; Wilms Tumor; Young Adult | 2017 |
Some GCR Polymorphisms (N363S, ER22/23EK, and Bcl-1) May Influence Steroid-induced Toxicities and Survival Rates in Children With ALL.
We investigated whether an altered individual glucocorticoid sensitivity due to particular glucocorticoid receptor single-nucleotide polymorphisms (SNPs) (N363S, ER22/23EK, and Bcl-1) influences the susceptibility to steroid-related toxicities, prognostic factors, and survival rates in children with acute lymphoblastic leukemia. In total, 346 pediatric patients with acute lymphoblastic leukemia were enrolled in our study. Their carrier status was investigated by allele-specific polymerase chain reaction analysis. Clinical and laboratory signs of glucocorticoid-related toxicities, day-8 prednisone response, 5-year event-free survival, and 5-year overall survival rates were analyzed and compared retrospectively. Hepatotoxicity occurred significantly more often in 363S carriers (P=0.004), and glucose metabolism abnormalities were more common in 363S carriers (P=0.001), but did not occur in patients with the ER22/23EK SNP. Hypertension and central nervous system/behavioral changes did not occur in patients with the ER22/23EK SNP. None of the patients with the N363S SNP, the ER22/23EK polymorphism, or the GG genotype for the Bcl-1 polymorphism had a poor prednisone response. The 363S carriers had significantly better 5-year event-free survival (P=0.012) and 5-year overall survival (P=0.013) rates compared with noncarriers. The Bcl-1 SNP was not associated with any of the toxicities investigated or survival. Children with the N363S polymorphism in the glucocorticoid receptor gene were more prone to steroid-related toxicities, whereas those with the ER22/23EK polymorphism were less susceptible. Children with the N363S polymorphism may have more favorable survival rates. Topics: Adolescent; Child; Child, Preschool; Cyclin D1; Disease-Free Survival; Glucocorticoids; Humans; Infant; Polymorphism, Genetic; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prednisone; Prognosis; Receptors, Glucocorticoid; Steroids; Survival Rate | 2016 |
Characterization of LEF1 High Expression and Novel Mutations in Adult Acute Lymphoblastic Leukemia.
Aberrant activation of the Wnt pathway plays a pathogenetic role in tumors and has been associated with adverse outcome in acute lymphoblastic leukemia (ALL). Lymphoid enhancer binding factor 1 (LEF1), a key mediator of Wnt signaling, has been linked to leukemic transformation, and LEF1 mutations have been identified in T-ALL. Here we found LEF1 is highly expressed in 25.0% adult ALL patients and LEF1 high expression was associated with high-risk leukemia factors (high WBC, Philadelphia chromosome positive, complex karyotype), shorter event-free survival (EFS), and high relapse rates in patients with B-ALL. LEF1 high expression is also associated with high mutation rate of Notch1 and JAK1 in T-ALL. We identified 2 novel LEF1 mutations (K86E and P106L) in 4 of 131 patients with ALL, and those patients with high-risk ALL (high WBC, complex karyotype). These results suggest a role for LEF1 mutations in leukemogenesis. We further explored the effect of the mutations on cell proliferation and found both mutations significantly promoted the proliferation of ALL cells. We also observed the effect of LEF1 and its mutations on the transcription of its targets, c-MYC and Cyclin D1. We found LEF1 increased the promoter activity of its targets c-MYC and Cyclin D1, and LEF1 K86E and P106L mutants further significantly enhanced this effect. We also observed that the c-MYC and Cyclin D1 mRNA levels were significantly increased in patients with LEF1 high expression compared with those with low expression. Taken together, our findings indicate high LEF1 expression and mutation are associated with high-risk leukemia and our results also revealed that LEF1 high expression and/or gain-of-function mutations are involved in leukemogenesis of ALL. Topics: Adolescent; Adult; Aged; Bone Marrow; Cell Line, Tumor; Cell Proliferation; Chromosome Aberrations; Cyclin D1; DNA Mutational Analysis; Female; Gene Expression Regulation, Leukemic; Humans; Lymphoid Enhancer-Binding Factor 1; Male; Middle Aged; Mutation; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Proto-Oncogene Proteins c-myc; Recurrence; Transcription, Genetic; Young Adult | 2015 |
CCND1 G870A polymorphism is associated with toxicity of methotrexate in childhood acute lymphoblastic leukemia.
CCND1 plays a key role in cell cycle progression and may cause methotrexate (MTX) resistance, as well as its cytotoxicity. CCND1 870A variant allele is associated with altered transcripts of this gene. We hypothesized that this polymorphism may contribute to the elimination rate and hepatotoxicity of MTX in childhood acute lymphoblastic leukemia (ALL). We genotyped the CCND1 G870A polymorphism in 125 childhood ALL patients treated with HDMTX. We found no notable associations between G870A polymorphism and the risk of delayed MTX elimination. However, this polymorphism was significantly associated with an increased risk of MTX hepatotoxicity [adjusted odds ratio (OR) = 4.44, 95% confidence interval (CI) = 1.35-14.63 for AG versus GG and adjusted OR = 6.39, 95% CI = 1.82-22.43 for AA versus GG]. Our results indicated that the CCND1 G870A polymorphism may be involved in the hepatotoxicity of MTX and act as a biological marker. Topics: Alleles; Amino Acid Substitution; Child; Child, Preschool; Cyclin D1; Drug Resistance, Neoplasm; Female; Genotype; Humans; Liver; Male; Methotrexate; Mutation; Odds Ratio; Polymorphism, Genetic; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2015 |
Aberrant DNA methylation and epigenetic inactivation of hMSH2 decrease overall survival of acute lymphoblastic leukemia patients via modulating cell cycle and apoptosis.
Altered regulation of many transcription factors has been shown to play important roles in the development of leukemia. hMSH2 can modulate the activity of some important transcription factors and is known to be a regulator of hematopoietic differentiation. Herein, we investigated epigenetic regulation of hMSH2 and its influence on cell growth and overall survival of acute lymphoblastic leukemia (ALL) patients.. hMSH2 promoter methylation status was assessed by COBRA and pyrosequencing in 60 ALL patients and 30 healthy volunteers. mRNA and protein expression levels of hMSH2, PCNA, CyclinD1, Bcl-2 and Bax were determined by real time PCR and Western blotting, respectively. The influence of hMSH2 on cell proliferation and survival was assessed in transient and stable expression systems.. mRNA and protein expression of hMSH2 and Bcl-2 was decreased, and that of PCNA, CyclinD1 and Bax was increased in ALL patients as compared to healthy volunteers (P<0.05). hMSH2 was inactivated in ALL patients through promoter hypermethylation. Furthermore, hMSH2 hypermethylation was found in relapsed ALL patients (85.7% of all cases). The median survival of patients with hMSH2 methylation was shorter than that of patients without hMSH2 methylation (log-rank test, P=0.0035). Over-expression of hMSH2 in cell lines resulted in a significant reduction in growth and induction of apoptosis.. This study suggests that aberrant DNA methylation and epigenetic inactivation of hMSH2 play an important role in the development of ALL through altering cell growth and survival. Topics: Adult; Apoptosis; bcl-2-Associated X Protein; Case-Control Studies; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Cyclin D1; DNA Methylation; Female; Gene Expression; Gene Silencing; Humans; Male; MutS Homolog 2 Protein; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proliferating Cell Nuclear Antigen; Promoter Regions, Genetic; Proto-Oncogene Proteins c-bcl-2; Recurrence; RNA, Messenger; Survival Rate | 2014 |
Farnesyltransferase inhibitors: molecular evidence of therapeutic efficacy in acute lymphoblastic leukemia through cyclin D1 inhibition.
Farnesyltransferase inhibitors have the ability to interfere with various intracellular pathways, reducing cell survival and proliferation. They have become an attractive tool for cancer therapy, namely acute leukemias. In this work, we have studied the efficacy of α-hydroxyfarnesylphosphonic acid (α-HFPA) in CEM (acute T-cell lymphoblastic leukemia) in culture.. CEM cells were incubated with α-HFPA at different concentrations; viability and proliferation studies were performed using the trypan blue exclusion assay and cell morphological analysis. Expression of lamin A/C, cyclin D1 and BAD were analyzed by flow cytometry.. Our results show that α-HFPA significantly decreases Farnesyltransferase activity, reduces cell proliferation and induces cell death through apoptosis in CEM cells, which is correlated with a reduction of cyclin D1 levels.. This study suggests that α-HFPA blocks the cell cycle and induces cell death through apoptosis in CEM cells and may be a therapeutic approach in ALL. Topics: Cell Line, Tumor; Cyclin D1; Enzyme Inhibitors; Farnesol; Farnesyltranstransferase; Humans; Organophosphonates; Phosphorous Acids; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2012 |
Strong lymphoid nuclear expression of SOX11 transcription factor defines lymphoblastic neoplasms, mantle cell lymphoma and Burkitt's lymphoma.
We surveyed lymphomas to determine the range of expression of the mantle cell lymphoma-associated SOX11 transcription factor and its relation to cyclin D1.. On hundred and seventy-two specimens were immunostained for the SOX11 N and C termini. Cyclin D1 was detected by immunohistochemistry and quantitative reverse transcriptase polymerase chain reaction; in situ hybridization for t(11;14) was applied when needed.. Nuclear SOX11 was strongly expressed in most B and T-lymphoblastic leukemia/lymphomas and half of childhood Burkitt's lymphomas, but only weakly expressed in some hairy cell leukemias. Chronic lymphocytic leukemia/lymphoma, marginal zone, follicular and diffuse large B-cell lymphomas were negative for SOX11, as were all cases of intermediate Burkitt's lymphomas/diffuse large B-cell lymphoma, myeloma, Hodgkin's lymphomas and mature T-cell and NK/T-cell lymphomas.. In addition to mantle cell lymphoma, SOX11 is strongly expressed only in lymphoblastic malignancies and Burkitt's lymphomas. Its expression is independent of cyclin D1 (except for weak expression in hairy cell leukemias) and unlikely to be due to translocations in lymphoid neoplasia. Topics: Burkitt Lymphoma; Cyclin D1; Gene Expression Regulation, Neoplastic; Humans; Immunohistochemistry; Lymphoma, Mantle-Cell; Polymerase Chain Reaction; Precursor Cell Lymphoblastic Leukemia-Lymphoma; SOXC Transcription Factors | 2009 |
Proangiogenic stimulation of bone marrow endothelium engages mTOR and is inhibited by simultaneous blockade of mTOR and NF-kappaB.
Most bone marrow (BM) malignancies develop in association with an angiogenic phenotype and increased numbers of endothelial cells. The molecular mechanisms involved in the modulation and recruitment of BM endothelium are largely unknown and may provide novel therapeutic targets for neoplastic diseases. We observed that angiogenic stimulation of BM endothelial cells activates mTOR and engages its downstream pathways 4E-BP1 and S6K1, which are inhibited by the mTOR-specific blockers rapamycin and CCI-779. Both mTOR blockers significantly inhibit growth factor- and leukemia-induced proliferation of BM endothelium by inducing G0/G1 cell-cycle arrest. This effect is associated with down-regulation of cyclin D1 and cdk2 phosphorylation, and up-regulation of the cdk inhibitors p27(kip1) and p21(cip1). Under conditions that reproduce the biomechanical fluidic environment of the BM, CCI-779 is equally effective in inhibiting BM endothelial-cell proliferation. Finally, simultaneous blockade of mTOR and NF-kappaB pathways synergize to significantly inhibit or abrogate the proliferative responses of BM endothelial cells to mitogenic stimuli. This study identifies mTOR as an important pathway for the proangiogenic stimulation of BM endothelium. Modulation of this pathway may serve as a valid therapeutic intervention in BM malignancies evolving in association with an angiogenic phenotype. Topics: Adult; Bone Marrow; Cell Cycle; Cell Proliferation; Cells, Cultured; Child; Cyclin D1; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinase Inhibitor p27; Endothelium; Gene Expression Regulation; Humans; Neovascularization, Pathologic; NF-kappa B; Phosphorylation; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Protein Kinases; TOR Serine-Threonine Kinases | 2006 |
Cyclin D1 gene polymorphism and susceptibility to childhood acute lymphoblastic leukemia in a Chinese population.
Cyclin D1 is a key protein involved in cell cycle regulation. A common A870G single nucleotide polymorphism in exon 4 of the cyclin D1 gene (CCND1) has an effect on the transcription of 2 different cyclin D1 messenger RNAs. Correlation between genetic polymorphism of A870G of CCND1 and clinical outcome among patients with acute lymphoblastic leukemia (ALL) has been reported. However, the effect on ALL occurrence remains unclear. To examine the genotypic frequency of CCND1 polymorphism, we performed a case-control study in a Chinese population of 183 children with ALL and 190 healthy controls. The genetic frequency of CCND1 had a significant overall correlation in patients and controls. The AA genotype of CCND1 showed a tendency to increase ALL risk 3.2898-fold compared with the AG + GG genotype (P = .0207). Stratification of patients according to cell type, risk level, and chemotherapeutic response showed significance for the AA genotype in T-cell ALL, ALL with high risk, and no complete remission (P = .047, P = .011, and P = .007, respectively). No gene dosage effect was observed in this study. The results of the present study suggested that CCND1 genetic polymorphism may be related to the occurrence of ALL in a population of Chinese children. Topics: Asian People; Child; China; Cyclin D1; Exons; Female; Genetic Predisposition to Disease; Genotype; Humans; Male; Point Mutation; Polymorphism, Single Nucleotide; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2005 |
Expression of cyclins D1, D2, and D3 and Ki-67 in Leukemia.
Cyclins are very important components of the cell cycle machinery because their levels regulate cell proliferation. They have also been found to be prognostic factors in various cancers. We studied the expression of the positive cell cycle regulators (D cyclins) and the cell proliferation marker (Ki-67) in human acute myeloid (AML), chronic myeloid (CML), acute lymphoblastic (ALL) and chronic lymphocytic (CLL) leukemia [mainly by comparative reverse transcription polymerase chain reaction (RT-PCR)]. Both leukemic and normal cells were positive for cyclin D3 expression. Significant differences were found in the expression of cyclin D1, which was the highest in leukocytes (CD19 + ) of CLL patients whereas lower expression was found in CML, AML and ALL patients and normal bone marrow and peripheral blood leukocytes (P < 0.001). The higher expression of cyclin D1 in leukocytes of CLL patients compared to CML patients was confirmed by quantitative real-time RT-PCR with a TaqMan probe in a subset of CLL and CML patients. Differences in cyclin D1 expression between CLL and CML patients were also confirmed on protein levels by western blotting. Expression of the proliferative marker Ki-67 was high in CML, ALL and AML cells and low in CD19-positive CLL cells. The results demonstrate that the level of cyclin D1 negatively correlates with the proliferation properties of leukemic cells. We did not find any significant relationship between cyclin D1 expression in cells of CML and AML patients and their clinical outcome. Topics: Acute Disease; Cell Proliferation; Cyclin D1; Cyclin D2; Cyclin D3; Cyclins; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Ki-67 Antigen; Leukemia; Leukemia, Lymphocytic, Chronic, B-Cell; Leukemia, Myelogenous, Chronic, BCR-ABL Positive; Leukemia, Myeloid; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Reverse Transcriptase Polymerase Chain Reaction | 2005 |
The spectrum of acute lymphoblastic leukemia with mature B-cell phenotype.
We showed heterogeneous disease spectrum among 15 acute lymphoblastic leukemia (ALL) cases with mature B-cell phenotype diagnosed over the past 7 years at our institution. Besides those with typical L3 morphology and 8q24 (c-myc) translocation (n=6), there were cases showing L1 or L2 morphology without 8q24 translocation (n=6), unusually large L3 blasts in hyperdiploid clone (n=2) and blastoid variant of mantle cell lymphoma (n=1). The expression of CD5 and cyclin D1 may need to be routinely determined on ALL cases with mature B-cell phenotype and non-L3 morphology to facilitate timely diagnosis of blastoid MCL and institution of suitable management. Topics: Adolescent; Adult; Aged; Aged, 80 and over; Bone Marrow Examination; Burkitt Lymphoma; CD5 Antigens; Child; Child, Preschool; Chromosomes, Human, Pair 14; Chromosomes, Human, Pair 8; Cyclin D1; Cytogenetic Analysis; Female; Humans; Immunophenotyping; Lymphoma, Mantle-Cell; Male; Middle Aged; Phenotype; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Retrospective Studies; Translocation, Genetic | 2003 |
Cell cycle alterations in the blastoid variant of mantle cell lymphoma (MCL-BV) as detected by gene expression profiling of mantle cell lymphoma (MCL) and MCL-BV.
Overexpression of cyclin D1 is necessary but by itself insufficient for the development of mantle cell lymphoma (MCL). To identify pathways in the pathogenesis of MCL and the blastoid variant (MLC-BV), we compared the gene-expression profiles of microdissected normal mantle cells, MCL, and MCL-BV by oligonucleotide microarrays and quantitative reverse transcriptase PCR (QRT-PCR). We identified and confirmed the overexpression of several genes in MCL-BV that are involved in the cell cycle control at the G1/S and G2/M checkpoints or inhibit apoptotic cell death. The highly expressed cyclin dependent kinase 4 (CDK4) is a cell cycle kinase that associates with cyclin D1 for the progression through the G1/S checkpoint, whereas overexpression of cdc28 protein kinase 1 (CKS1) blocks the inhibition of the cyclin D1/CDK4 complex by the CDK inhibitor p27/Kip1. Other highly expressed genes in MCL-BV that promote the cells through the G1/S-checkpoint include the oncogenes B-Myb, PIM1, and PIM2, and passage through the G2/M-checkpoint is enhanced by high levels of cdc25B. Furthermore, two highly expressed genes that inhibit apoptosis are defender against cell death (DAD1) and RSK1. In summary, our microarray and QRT-PCR analyses identified several candidate genes whose expression increased when comparing normal follicular mantles with MCL and MCLBV, suggesting a potential pathogenic role in the evolution of MCL-BV. Topics: Cell Cycle; Cell Transformation, Neoplastic; Cyclin D1; DNA, Neoplasm; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Lymphoma, Mantle-Cell; Neoplastic Stem Cells; Oligonucleotide Array Sequence Analysis; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger | 2003 |
The influence of cyclin D1 (CCND1) 870A>G polymorphism and CCND1-thymidylate synthase (TS) gene-gene interaction on the outcome of childhood acute lymphoblastic leukaemia.
The 870A>G polymorphism in the cyclin D1 (CCND1) gene modulates mRNA splicing, leading to altered protein that may affect the regulation of the G1/S cell-cycle checkpoint. This polymorphism has been reported to influence susceptibility to and progression of several malignancies. Furthermore, the change of retinoblastoma protein regulation mediated by CCND1 may play a role in the development of methotrexate (MTX) resistance via an associated higher activity of enzymes that are inhibited by MTX. This study shows that children with acute lymphoblastic leukaemia (ALL) who are homozygous for the CCND1 A variant have a lower probability of event-free survival (P = 0.006) compared to carriers of the G variant. A significant result is retained in the presence of other prognostic factors. This impact is even more apparent in individuals who are also homozygous for thymidylate synthase (TS) triple repeat (P < 0.00005), which has previously been shown to influence the outcome of childhood ALL. Topics: Child; Cyclin D1; France; Genetic Variation; Heterozygote; Homozygote; Humans; Polymorphism, Genetic; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Prognosis; Risk Factors; Thymidylate Synthase; Trinucleotide Repeats | 2003 |
BCL-1 rearrangement in acute lymphocytic leukemia and its clinical significance.
BCL-1 rearrangement (BCL-1/IgH gene rearrangement) in acute lymphocytic leukemia and its clinical significance was investigated. In 38 patients with acute lymphocytic leukemia (ALL), the genomic DNA of mononuclear cells isolated from peripheral blood and bone marrow was amplified by using hemi-nested polymerase chain reaction (PCR) technique and the expression of cyclin D1 protein of mononuclear cells was detected by using immunohistochemical method. Ten patients with acute granulocytic leukemia, 2 with chronic granulocytic leukemia and 10 with normal bone marrow served as control group. The results showed that BCL-1 rearrangement was detectable in 3 of 38 ALL patients (7.9%) and cyclin D1 protein positive expression was detected in 4 ALL patients (10.5%). Three ALL patients with BCL-1 rearrangement were all B-cell leukemia (B-ALL) and accompanied by cyclin D1 protein expression. No BCL-1/IgH rearrangement or cyclin D1 protein expression was detected in 12 patients with granulocytic leukemia and 10 cases of normal bone marrow. Leukocyte counts in peripheral blood of B-ALL patients with BCL-1 rearrangement and (or) cyclin D1 protein expression were significantly increased and the patients had bad reaction to chemotherapy. It was concluded that: 1) BCL-1/IgH gene rearrangement were detected in acute B lymphocytic leukemia; 2) B-ALL patients with BCL-1 rearrangement and (or) cyclin D1 protein expression had poor prognosis. Topics: Adolescent; Adult; Burkitt Lymphoma; Child; Cyclin D1; Female; Gene Rearrangement; Genes, bcl-1; Humans; Immunoglobulin Heavy Chains; Male; Middle Aged; Phenotype; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 2001 |
Immunohistochemical expression of the p53, mdm2, p21/Waf-1, Rb, p16, Ki67, cyclin D1, cyclin A and cyclin B1 proteins and apoptotic index in T-cell lymphomas.
Fifty-seven cases of T-cell lymphomas (TCL) including 5 lymphoblastic (T-LBL) and 52 peripheral TCL (PTCL) were analyzed by immunohistochemistry for the expression of p53, mdm2, p21, Rb, cyclin D1, cyclin A, cyclin B1, and Ki67/MIB1 proteins and 39/52 PTCL were also analyzed for the expression of p16 protein and for the presence of apoptotic cells by the TUNEL method. The aim was to search for abnormal immunoprofiles of p53 and Rb growth control pathways and to determine the proliferative activity and the apoptotic index of TCL. Abnormal overexpression of p53, p21 and mdm2, in comparison to normal lymph nodes, was found in 12/57, 10/57 and 2/57 cases of TCL, respectively. Abnormal loss of Rb and p16 expression was found in 1/57 and 2/39 cases, respectively, whereas abnormal overexpression of cyclin D1 was not detected in any of the 57 cases. Our data revealed entity-related p53/p21/mdm2 phenotypes. Indeed, most nodal and cutaneous CD30+ anaplastic large cell lymphomas (ALCL) showed concomitant overexpression of p53 and p21 proteins (7/8 cases), and mdm2 was overexpressed in 2 p53-positive nodal ALCL. In contrast, overexpression of p53 was found in 3/17 cases of nodal peripheral TCL unspecified (PTCL-UC) and 2/7 non-ALCL cutaneous pleomorphic TCL. Overexpression of p21 protein was detected in 2/3 p53-positive PTCL-UC and in 1/2 p53-positive non-ALCL cutaneous pleomorphic TCL. Finally, all the remaining 25 cases of TCL did not show p53 and p21 overexpression. Overall, the p53+/p21+ phenotype in 10/57 TCL suggests wild-type p53 capable of inducing p21 expression. The highest apoptotic index (AI) was found in ALCL and a positive correlation between apoptotic index and Ki67 index (p<0.001) was detected. Ki67, cyclin A and cyclin B1 expression was found in all 57 TCL and on the basis of the combined use of these 3 variables, 3 groups of proliferative activity could be determined: a) high in ALCL and T-LBL, b) low in mycosis fungoides (MF) and gammadelta hepatosplenic TCL, and c) intermediate in the remaining TCL entities. The proliferative activity in the 12 p53 overexpressing cases was higher in comparison to the 45 p53-negative cases. Ki67 expresion in more than 25% of tumour cells showed significant correlation with p53 overexpression (p<0.001). Rb expression tended to be parallel to Ki67, cyclin A and cyclin B1 expression in all but one case of nodal PTCL-UC which displayed loss of RB expression. Interestingly, this case was p53-negative, whereas the p53-po Topics: Adenovirus E1A Proteins; Apoptosis; Carrier Proteins; Cell Cycle Proteins; Cyclin A; Cyclin B; Cyclin B1; Cyclin D1; Cyclin-Dependent Kinase Inhibitor p16; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Humans; Immunohistochemistry; In Situ Nick-End Labeling; Ki-67 Antigen; Lymphoma, T-Cell, Peripheral; Neoplasm Proteins; Nuclear Proteins; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-mdm2; Repressor Proteins; Statistics as Topic; Tumor Suppressor Protein p53 | 2001 |
Hypermethylation of p16 and p15 genes and RB protein expression in acute leukemia.
Both p16 and p15, encoded by genes located on chromosome 9p21, are inhibitors of cyclin-dependent kinases 4/6 (CDK4/6) and upstream regulators of RB function, and set up the RB/p16 tumor suppressive pathway, which is abrogated frequently in human neoplasms, either through inactivation of the RB or p16 tumor-suppressor protein, or alteration of the cyclin D1 or CDK4 oncoproteins. In hematological malignancies, deletion of p16/p15 locus has been shown to be highly specific to lymphoid malignancies, and more particularly to T-cell acute lymphoblastic leukemia (T-ALL). However, in the other subsets of ALL, deletions of p16 and p15 are relatively rare events. To investigate whether these genes are inactivated by methylation of the 5' CpG islands, we examined 35 leukemia cell lines and 29 childhood acute myeloid leukemia (AML) patients by Southern blot, polymerase chain reaction (PCR) and Western blot analyses. We found methylation of p16 in 12 (50%) of 24 ALL cell lines, 5 (50%) of 10 AML cell lines without homozygous deletion of p16, and 11 (38%) of 29 AML patients. Those leukemia cell lines subjected to p16 methylation were found to have lost p16 protein expression. The p15 gene was methylated in 10 (34%) of 29 ALL cell lines, 6 (60%) of 10 AML cell lines without homozygous deletion of p15, and 15 (52%) of 29 AML patients. These results revealed the frequent methylation of p16 and p15 genes in B-ALL and AML despite a low frequency of p16 and p15 deletions and mutations in these leukemias. In the study for expression of RB protein, we found no expression of RB in 4 of 16 leukemia cell lines. Inactivation of the p16 gene was found in all the cell lines with expression of RB. Neither amplification nor rearrangement of cyclin D1 gene was found in any cell lines. These results suggest that inactivation of p16 and p15 genes is one of the most common genetic events in acute leukemia, and plays an important role for the RB/p16 pathway in the pathogenesis of acute leukemia. Topics: Acute Disease; Burkitt Lymphoma; Carrier Proteins; Cell Cycle Proteins; CpG Islands; Cyclin D1; Cyclin-Dependent Kinase Inhibitor p15; Cyclin-Dependent Kinase Inhibitor p16; DNA Methylation; DNA, Neoplasm; Gene Expression Regulation, Leukemic; Genes, p16; Genes, Retinoblastoma; Humans; Leukemia-Lymphoma, Adult T-Cell; Leukemia, Myeloid; Loss of Heterozygosity; Molecular Probe Techniques; Neoplasm Proteins; Polymerase Chain Reaction; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Retinoblastoma Protein; Sequence Deletion; Tumor Cells, Cultured; Tumor Suppressor Proteins | 2000 |
[Expression and role of cyclin D3 in childhood acute leukemia].
To investigate the expression and role of cyclin D in childhood acute leukemia(AL).. Immunohistochemistry was used to detect the expression of cyclin D1, D2, D3 in 43 samples of childhood AL patients and three leukemic cell lines. Cyclin D3 antisense oligodeoxynucleotides were used in in vitro culture study.. Cyclin D3 expression was positive in 47% (14/30) of the childhood acute lymphoblastic leukemia and 38% (5/13) of the acute myelogenous leukemia patients and in CEM cells. Cyclin D1 was overexpressed in 5% (2/43) of the AL patients. No overexpression of cyclin D2 was detected. Incubation of CEM cells with cyclin D3 antisense oligodeoxynucleotides resulted in inhibition of cell proliferation.. Cyclin D3 was overexpressed in childhood AL and it played an important role in the proliferation of leukemic cells. Topics: Adolescent; Cell Division; Child; Child, Preschool; Cyclin D1; Cyclin D2; Cyclin D3; Cyclins; Humans; Infant; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Tumor Cells, Cultured | 1999 |
[Expression of cyclin D1 mRNA in acute leukemia patients and its clinical implication].
To determine the expression of cyclin D1 gene in patients with acute leukemia and evaluate the relationship between cyclin D1 expression and treatment outcomes.. The expression of cyclin D1 mRNA was determined in bone marrow cells from 65 acute leukemia patients and 5 normal subjects by RT-PCR technique.. Cyclin D1 mRNA was negative in normal bone marrow cells. Among leukemia patients, the rate of cyclin D1 expression was 26.2%, and the highest(63.6%) rate was in the relapse group and the lowest(8.3%) in the CR group. Relapse rate of the patients with cyclin D1 expression was higher than that of those with negative expression over 1-13 months follow-up.. Overexpression of cyclin D1 does exist in acute leukemia patients and is correlated with the disease progression, especially with relapse. Topics: Adolescent; Adult; Aged; Bone Marrow Cells; Cyclin D1; Female; Humans; Leukemia, Myeloid, Acute; Male; Middle Aged; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Recurrence; RNA, Messenger | 1999 |
Cyclin D1 overexpression detected by a simple competitive reverse transcription-polymerase chain reaction assay for lymphoid malignancies.
t(11;14)(q13;q32) observed in B-cell malignancies is associated with cyclin D1 (bcl-1, PRAD1, CCND1) overexpression. We devised a simple competitive reverse transcription-polymerase chain reaction (RT-PCR) assay for rapid detection of cyclin D1 overexpression. Sharing a single upstream primer derived from a homologous sequence in cyclins D1, D2 and D3, each PCR product serves as a competitor and cyclin D1 overexpression is determined by comparing the intensities of the three amplified products. We analyzed cyclin D1 in clinical specimens from 104 patients with lymphoid malignancies. Cyclin D1 overexpression was evident in 13 of 104 (7/72 non-Hodgkin's lymphomas, 0/6 adult T-cell lymphoma/leukemias, 0/4 Hodgkin's diseases, 0/11 acute lymphoblastic leukemias, 3/4 multiple myelomas, 1/2 Waldenström's macroglobulinemias, 1/2 prolymphocytic leukemias and 1/3 chronic lymphocytic leukemias). Among 72 patients for whom cytogenetic studies had been done, all 7 patients with t(11;14) were positive. The relative expression levels of D-type cyclins altered dramatically in the presence of t(11;14). Thus, this RT-PCR assay can identify tumors with cyclin D1 overexpression. Cyclin D1 overexpression was frequent in extranodal specimens (11 out of 32 vs. 2 of 72 lymph nodes) and was restricted to specific types of lymphoid malignancies, as observed using other methods. This reliable assay should be suitable to provide clinical guidance for the diagnosis and management of lymphoid malignancies, especially in the case of extranodal involvement. Topics: Adolescent; Adult; Aged; Aged, 80 and over; Bone Marrow; Cyclin D1; Female; Hodgkin Disease; Humans; Leukemia-Lymphoma, Adult T-Cell; Leukemia, Lymphocytic, Chronic, B-Cell; Lymphoma, Non-Hodgkin; Lymphoproliferative Disorders; Male; Middle Aged; Multiple Myeloma; Polymerase Chain Reaction; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Transcription, Genetic; Waldenstrom Macroglobulinemia | 1998 |
BCL-1 (PRAD-1/cyclin D-1) overexpression distinguishes the blastoid variant of mantle cell lymphoma from B-lineage lymphoblastic lymphoma.
The blastoid variant of mantle cell lymphoma (MCL-BV) can occur de novo or can represent a morphologic transformation of MCL associated with aggressive clinical disease. Its cytologic appearance is very similar to that of lymphoblastic lymphoma (LBL) because of its characteristic nuclear features and high proliferative rate. To assess the usefulness of antibodies to cyclin D-1 (BCL-1/ PRAD-1), CD99 (12E7), CD34, and TdT in distinguishing between MCL-BV and LBL in formalin-fixed, paraffin-embedded tissue, we studied from the Stanford data base 10 cases originally diagnosed as B-lineage LBL, 5 MCL-BVs, 2 cases thought likely to represent MCL-BV, and 2 blastic lymphomas whose morphology and immunophenotype were indeterminate. Six (60%) of 10 LBLs stained with CD99, as opposed to none of 7 MCL-BVs. Four (40%) of 10 LBLs reacted with CD34, as compared with none of 7 MCL-BVs. Eight (89%) of nine LBLs were positive for TdT, but all of the four MCL-BVs tested were negative. In contrast, the anti-cyclin D-1 antibody stained the nuclei of all of the MCL-BVs and none of the LBLs tested. On the basis of our evaluation, the probable MCL-BV cases were considered to be definite MCL-BV. Of the indeterminate cases, one was considered to be LBL, whereas we felt that the other represented MCL-BV. We conclude that staining formalin-fixed, paraffin-embedded, high-grade lymphomas with anti-cyclin D-1 antibody is useful in confirming the diagnosis of MCL-BV, whereas positive reactions with CD99, CD34, and particularly TdT are more characteristic of LBL. Topics: 12E7 Antigen; Adult; Aged; Antigens, CD; Antigens, CD34; Biomarkers, Tumor; Cell Adhesion Molecules; Child; Cyclin D1; Cyclins; Diagnosis, Differential; DNA Nucleotidylexotransferase; Female; Humans; Immunohistochemistry; Immunophenotyping; Lymphoma, B-Cell; Lymphoma, Non-Hodgkin; Male; Middle Aged; Oncogene Proteins; Precursor Cell Lymphoblastic Leukemia-Lymphoma | 1997 |
Fluorescent in situ hybridization analysis of an atypical t(11;14)(q13;q32) without expression of the cyclin D1 gene.
Topics: Adult; Chromosomes, Human, Pair 11; Chromosomes, Human, Pair 14; Cyclin D1; Cyclins; Female; Gene Expression; Humans; In Situ Hybridization, Fluorescence; Oncogene Proteins; Oncogenes; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Translocation, Genetic | 1993 |