Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Dermatopathol ; 41(10): 757-766, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30973519

ABSTRACT

INTRODUCTION: Cutaneous composite lymphoma (CCL) is extremely rare. When 2 potentially distinct lymphoid lesions occur at one skin site, distinguishing between one neoplastic clone and a secondary reactionary lymphoid response versus a second neoplasm is difficult. In this study, we describe a unique case of CCL along with a review of reported cases in literature to identify clues and discuss issues that are relevant to the diagnosis of CCL. DESIGN: Review of a CCL case from our institution and a systematic review of reported cases of CCL in the literature. RESULTS: A total of 18 studies describing 22 cases and a case report from our institution are included. The mean age at diagnosis was 68 years. Most cases herein presented with multiple skin lesions (67%) and reported a history of immune suppression (76%). Nineteen cases (83%) had a combination of T-cell and B-cell neoplasms, whereas the remaining cases had 2 distinct B-cell clones. Clonal differentiation was confirmed based on morphology and immunohistochemistry in all cases, and by polymerase chain reaction studies in 19 cases. Complete remission was achieved in only one quarter of reported cases. CONCLUSION: Diagnosing CCL can be challenging because accurate differentiation of 2 or more clonal populations at 1 site is tedious. A stepwise approach and integration of clinical, morphologic, immunohistochemistry, and molecular data along with an understanding of the prognosis of the lymphomas in question is essential for an accurate diagnosis and necessary because of therapeutic and prognostic implications.


Subject(s)
Composite Lymphoma/diagnosis , Skin Neoplasms/diagnosis , Female , Humans , Middle Aged
2.
Proc Natl Acad Sci U S A ; 112(46): E6359-68, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578796

ABSTRACT

Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


Subject(s)
Erythroblasts/metabolism , GTPase-Activating Proteins/biosynthesis , Gene Expression Regulation , Myelodysplastic Syndromes/metabolism , Actins/genetics , Actins/metabolism , Animals , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Erythroblasts/pathology , Female , GTPase-Activating Proteins/genetics , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
3.
Blood ; 123(2): 228-38, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24264229

ABSTRACT

Therapy-related myeloid neoplasms (t-MN) are a late complication of the successful use of cytotoxic therapy for patients with cancer. A heterozygous deletions of the long arm of chromosome 5 [del(5q)], observed in 40% of patients, is associated with prior exposure to alkylating agents, and a high frequency of TP53 loss or mutation. In previous studies, we demonstrated that haploinsufficiency of 2 del(5q) genes, Egr1, and Apc, individually play a role in the pathogenesis of hematologic disease in mice. We now show that loss of one copy of Egr1 or Tp53 in an Apc haploinsufficient background (Apc (del/+)) accelerated the development of a macrocytic anemia with monocytosis, early features of t-MN. The development of anemia was significantly accelerated by treatment of mice with the alkylating agent, N-ethyl-N-nitrosourea (ENU), regardless of the levels of expression of Egr1 and Tp53. Transplantation of either wild type; Egr1(+/-); Tp53(+/-); Apc(del/+); or Egr1(+/-), Apc(del/+) bone marrow cells into lethally irradiated Apc(del/+) recipients resulted in rapid development of anemia that was further accelerated by administration of ENU to recipients, demonstrating that the Apc(del/+)-induced anemia was cell extrinsic and potentiated by ENU mutagenesis. These data emphasize the synergistic role of cell intrinsic and cell extrinsic (microenvironment) factors in the pathogenesis of t-MN, and raise awareness of the deleterious effects of cytotoxic therapy on the stromal microenvironment.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5 , Early Growth Response Protein 1/genetics , Haploinsufficiency , Tumor Suppressor Protein p53/genetics , Alleles , Anemia, Macrocytic/chemically induced , Anemia, Macrocytic/genetics , Anemia, Macrocytic/mortality , Animals , Apoptosis/genetics , Bone Marrow/drug effects , Cellular Microenvironment/drug effects , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis/genetics , Ethylnitrosourea/adverse effects , Genes, Lethal , Genotype , Heterozygote , Humans , Mice , Mice, Transgenic , Spleen/metabolism , Spleen/pathology
4.
Blood ; 123(7): 1069-78, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24381225

ABSTRACT

An interstitial deletion of chromosome 5, del(5q), is the most common structural abnormality in primary myelodysplastic syndromes (MDS) and therapy-related myeloid neoplasms (t-MNs) after cytotoxic therapy. Loss of TP53 activity, through mutation or deletion, is highly associated with t-MNs with a del(5q). We previously demonstrated that haploinsufficiency of Egr1 and Apc, 2 genes lost in the 5q deletion, are key players in the progression of MDS with a del(5q). Using genetically engineered mice, we now show that reduction or loss of Tp53 expression, in combination with Egr1 haploinsufficiency, increased the rate of development of hematologic neoplasms and influenced the disease spectrum, but did not lead to overt myeloid leukemia, suggesting that altered function of additional gene(s) on 5q are likely required for myeloid leukemia development. Next, we demonstrated that cell intrinsic loss of Tp53 in hematopoietic stem and progenitor cells haploinsufficient for both Egr1 and Apc led to the development of acute myeloid leukemia (AML) in 17% of mice. The long latency (234-299 days) and clonal chromosomal abnormalities in the AMLs suggest that additional genetic changes may be required for full transformation. Thus, loss of Tp53 activity in cooperation with Egr1 and Apc haploinsufficiency creates an environment that is permissive for malignant transformation and the development of AML.


Subject(s)
Chromosome Deletion , Early Growth Response Protein 1/genetics , Genes, APC , Genes, p53/physiology , Haploinsufficiency , Leukemia, Myeloid, Acute/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Stem Cells ; 33(12): 3643-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26417967

ABSTRACT

Epigenetic alterations play a central role in the control of normal and malignant blood cell development. We demonstrate here that expression of a truncated DNA methyltransferase 3B isoform DNMT3B7, which has been shown to alter cellular epigenetic patterns, decreases the overall number of hematopoietic stem and progenitor cells (HSPCs), and markedly diminishes blood cell reconstitution within the female hormonal microenvironment. Gene expression profiling of HSPCs isolated from DNMT3B7 transgenic embryos identified Apolipoprotein E (Apoe) as overexpressed. The CpG island controlling Apoe expression had lower levels of modified cytosines in DNMT3B7 transgenic HSPCs, corresponding with the observed increase in gene expression. Furthermore, we observed that spleens and bone marrows of female mice transplanted with DNMT3B7 transgenic HSPCs express very high levels of Apoe. Finally, the introduction of Apoe-overexpressing HSPCs into male recipients decreased bone marrow engraftment, recapitulating our original observations in female recipients. Our work reveals a dynamic interplay between the intrinsic epigenetic changes in HSPCs and extrinsic endocrine factors acting on these cells to regulate the efficiency of HSPC engraftment and reconstitution. We have identified a novel mechanism by which gender-specific hormones modulate HSPC function, which could serve as a target for augmenting hematopoiesis in cases with limited HSC functionality.


Subject(s)
Apolipoproteins E/biosynthesis , CpG Islands/physiology , Epigenesis, Genetic/physiology , Hematopoiesis/physiology , Sex Characteristics , Animals , Apolipoproteins E/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Male , Mice , Mice, Knockout
6.
Blood ; 121(11): 2059-63, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23315164

ABSTRACT

The drivers of abnormal DNA methylation in human cancers include widespread aberrant splicing of the DNMT3B gene, producing abnormal transcripts that encode truncated proteins that may act as dominant negative isoforms. To test whether reduced Dnmt3b dosage can alter tumorigenesis, we bred Dnmt3b(+/-) mice to Eµ-Myc mice, a mouse model susceptible to B-cell lymphomas. Eµ-Myc/Dnmt3b(+/-) mice showed a dramatic acceleration of lymphomagenesis, greater even than that observed in Eµ-Myc mice that express a truncated DNMT3B isoform found in human tumors, DNMT3B7. This finding indicates that Dnmt3b can act as a haploinsufficient tumor suppressor gene. Although reduction in both Dnmt3b dosage and expression of DNMT3B7 within the Eµ-Myc system had similar effects on tumorigenesis and DNA hypermethylation, different molecular mechanisms appear to underlie these changes. This study offers insight into how de novo DNA methyltransferases function as tumor suppressors and the sensitivity of Myc-induced lymphomas to DNA methylation.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , Genes, Tumor Suppressor/physiology , Haploinsufficiency/physiology , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-myc/physiology , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Haploinsufficiency/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Promoter Regions, Genetic/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , DNA Methyltransferase 3B
7.
Blood ; 121(6): 975-83, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23212519

ABSTRACT

Loss of chromosome 7 and del(7q) [-7/del(7q)] are recurring cytogenetic abnormalities in hematologic malignancies, including acute myeloid leukemia and therapy-related myeloid neoplasms, and associated with an adverse prognosis. Despite intensive effort by many laboratories, the putative myeloid tumor suppressor(s) on chromosome 7 has not yet been identified.We performed transcriptome sequencing and SNP array analysis on de novo and therapy-related myeloid neoplasms, half with -7/del(7q). We identified a 2.17-Mb commonly deleted segment on chromosome band 7q22.1 containing CUX1, a gene encoding a homeodomain-containing transcription factor. In 1 case, CUX1 was disrupted by a translocation, resulting in a loss-of-function RNA fusion transcript. CUX1 was the most significantly differentially expressed gene within the commonly deleted segment and was expressed at haploinsufficient levels in -7/del(7q) leukemias. Haploinsufficiency of the highly conserved ortholog, cut, led to hemocyte overgrowth and tumor formation in Drosophila melanogaster. Similarly, haploinsufficiency of CUX1 gave human hematopoietic cells a significant engraftment advantage on transplantation into immunodeficient mice. Within the RNA-sequencing data, we identified a CUX1-associated cell cycle transcriptional gene signature, suggesting that CUX1 exerts tumor suppressor activity by regulating proliferative genes. These data identify CUX1 as a conserved, haploinsufficient tumor suppressor frequently deleted in myeloid neoplasms.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Acute Disease , Animals , Blotting, Western , Cell Line, Tumor , Drosophila melanogaster/genetics , Gene Expression Profiling , Haploinsufficiency , HeLa Cells , Homeodomain Proteins/metabolism , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , K562 Cells , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nuclear Proteins/metabolism , RNA Interference , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Translocation, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , U937 Cells , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 109(15): 5735-9, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22451912

ABSTRACT

Diffuse large B-cell lymphomas in humans are associated with chromosomal rearrangements (∼40%) and/or mutations disrupting autoregulation (∼16%) involving the BCL6 gene. Studies of lymphoma development in humans and mouse models have indicated that lymphomagenesis evolves through the accumulation of multiple genetic alterations. Based on our prior studies, which indicated that carcinogen-induced DNA mutations enhance the incidence of lymphomas in our mouse model expressing a human BCL6 transgene, we hypothesized that mutated genes are likely to play an important cooperative role in BCL6-associated lymphoma development. We used retroviral insertional mutagenesis in an effort to identify which genes cooperate with BCL6 in lymphomagenesis in our BCL6 transgenic mice. We identified PIM1 as the most frequently recurring cooperating gene in our murine BCL6-associated lymphomas (T- and B-cell types), and we observed elevated levels of PIM1 mRNA and protein expression in these neoplasms. Further, immunohistochemical staining, which was performed in 20 randomly selected BCL6-positive human B- and T-cell lymphomas, revealed concurrent expression of BCL6 and PIM1 in these neoplasms. As PIM1 encodes a serine/threonine kinase, PIM1 kinase inhibition may be a promising therapy for BCL6/PIM1-positive human lymphomas.


Subject(s)
DNA-Binding Proteins/genetics , Lymphoma/genetics , Lymphoma/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Proteins c-pim-1/genetics , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Mutagenesis, Insertional/genetics , Proto-Oncogene Proteins c-bcl-6 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae/genetics , Survival Analysis
9.
Br J Haematol ; 166(4): 550-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24931631

ABSTRACT

-7/del(7q) occurs in half of myeloid malignancies with adverse-risk cytogenetic features and is associated with poor survival. We identified the spectrum of mutations that co-occur with -7/del(7q) in 40 patients with de novo or therapy-related myeloid neoplasms. -7/del(7q) leukaemias have a distinct mutational profile characterized by low frequencies of alterations in genes encoding transcription factors, cohesin and DNA-methylation-related proteins. In contrast, RAS pathway activating mutations occurred in 50% of cases, a significantly higher frequency than other acute myeloid leukaemias and higher than previously reported. Our data provide guidance for which pathways may be most relevant in the treatment of adverse-risk myeloid leukaemia.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Leukemia, Myeloid, Acute/genetics , Homeodomain Proteins/genetics , Humans , Nuclear Proteins/genetics , Repressor Proteins/genetics , Risk Factors , Transcription Factors
10.
Blood Cells Mol Dis ; 52(1): 68-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23910958

ABSTRACT

The BCL6 gene, which is expressed in certain B- and T-cell human lymphomas, is involved with chromosomal rearrangements and mutations in a number of these neoplasms. Lymphomagenesis is believed to evolve through a multi-step accumulation of genetic alterations in these tumors. We used retroviral insertional mutagenesis in transgenic mice expressing the human BCL6 transgene in order to identify genes that cooperate with BCL6 during lymphomatous transformation. We previously reported PIM1 as the most frequently recurring cooperating gene in this model. We now report three newly identified cooperating genes-GFI1B, EVI5, and MYB-that we identified in the lymphomas of retroviral-injected BCL6 transgenic mice (but not in retroviral-injected non-transgenic controls); mRNA and protein expression of GFI1B and EVI5 were decreased in the murine tumors, whereas MYB mRNA and protein expression were increased or decreased. These findings correlated with protein expression in human lymphomas, both B- and T-cell. Improved therapy of lymphomas may necessitate the development of combinations of drugs that target the alterations specific to each neoplasm.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Lymphoma, T-Cell/genetics , Oncogene Proteins v-myb/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/metabolism , Female , GTPase-Activating Proteins , Genetic Vectors , Humans , Immunohistochemistry , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Mice , Mice, Transgenic , Oncogene Proteins v-myb/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-6 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Retroviridae/genetics , Signal Transduction , Transcription Factors/metabolism
11.
Haematologica ; 99(5): 821-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24463215

ABSTRACT

Acute myeloid leukemia and myelodysplastic syndrome with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) have a poor prognosis. Indeed, the inv(3)(q21q26.2)/t(3;3)(q21;q26.2) has been recognized as a poor risk karyotype in the revised International Prognostic Scoring System. However, inv(3)(q21q26.2)/t(3;3)(q21;q26.2) is not among the cytogenetic abnormalities pathognomonic for diagnosis of acute myeloid leukemia irrespective of blast percentage in the 2008 WHO classification. This multicenter study evaluated the clinico-pathological features of acute myeloid leukemia/myelodysplastic syndrome patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) and applied the revised International Prognostic Scoring System to myelodysplastic syndrome patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2). A total of 103 inv(3)(q21q26.2)/t(3;3)(q21;q26.2) patients were reviewed and had a median bone marrow blast count of 4% in myelodysplastic syndrome (n=40) and 52% in acute myeloid leukemia (n=63) (P<0.001). Ninety-one percent of patients showed characteristic dysmegakaryopoiesis. There was no difference in overall survival between acute myeloid leukemia and myelodysplastic syndrome patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) (12.9 vs. 7.9 months; P=0.16). Eighty-three percent of patients died (median follow up 7.9 months). Complex karyotype, monosomal karyotype and dysgranulopoiesis (but not blast percentage) were independent poor prognostic factors in the entire cohort on multivariable analysis. The revised International Prognostic Scoring System better reflected overall survival of inv(3)(q21q26.2)/t(3;3)(q21;q26.2) than the International Prognostic Scoring System but did not fully reflect the generally dismal prognosis. Our data support consideration of myelodysplastic syndrome with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) as an acute myeloid leukemia with recurrent genetic abnormalities, irrespective of blast percentage.


Subject(s)
Abnormal Karyotype , Bone Marrow/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosome Inversion , Chromosomes, Human, Pair 3 , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Patient Outcome Assessment , Prognosis , Translocation, Genetic , Young Adult
12.
Nat Genet ; 32(1): 148-52, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12172547

ABSTRACT

Children with Down syndrome have a 10-20-fold elevated risk of developing leukemia, particularly acute megakaryoblastic leukemia (AMKL). While a subset of pediatric AMKLs is associated with the 1;22 translocation and expression of a mutant fusion protein, the genetic alterations that promote Down syndrome-related AMKL (DS-AMKL) have remained elusive. Here we show that leukemic cells from every individual with DS-AMKL that we examined contain mutations in GATA1, encoding the essential hematopoietic transcription factor GATA1 (GATA binding protein 1 or globin transcription factor 1). Each mutation results in the introduction of a premature stop codon in the gene sequence that encodes the amino-terminal activation domain. These mutations prevent synthesis of full-length GATA1, but not synthesis of a shorter variant that is initiated downstream. We show that the shorter GATA1 protein, which lacks the N-terminal activation domain, binds DNA and interacts with its essential cofactor Friend of GATA1 (FOG1; encoded by ZFPM1) to the same extent as does full-length GATA1, but has a reduced transactivation potential. Although some reports suggest that the activation domain is dispensable in cell-culture models of hematopoiesis, one study has shown that it is required for normal development in vivo. Together, these findings indicate that loss of wildtype GATA1 constitutes one step in the pathogenesis of AMKL in Down syndrome.


Subject(s)
DNA-Binding Proteins/genetics , Down Syndrome/complications , Down Syndrome/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Mutation , Proto-Oncogene Proteins , Transcription Factors/genetics , Carrier Proteins/metabolism , Child, Preschool , Core Binding Factor Alpha 2 Subunit , DNA/metabolism , Erythroid-Specific DNA-Binding Factors , Female , GATA1 Transcription Factor , Genetic Predisposition to Disease , Humans , Infant , Leukemia, Megakaryoblastic, Acute/etiology , Male , Nuclear Proteins/metabolism , Polymorphism, Single-Stranded Conformational , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Tumor Cells, Cultured
13.
Blood ; 115(17): 3481-8, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20065296

ABSTRACT

Loss of a whole chromosome 5 or a deletion of the long arm of chromosome 5, -5/del(5q), is a recurring abnormality in myeloid neoplasms. The APC gene is located at chromosome band 5q23, and is deleted in more than 95% of patients with a -5/del(5q), raising the question of whether haploinsufficiency of APC contributes to the development of myeloid neoplasms with loss of 5q. We show that conditional inactivation of a single allele of Apc in mice leads to the development of severe anemia with macrocytosis and monocytosis. Further characterization of the erythroid lineage revealed that erythropoiesis is blocked at the early stages of differentiation. The long-term hematopoietic stem cell (LT-HSC) and short-term HSC (ST-HSC) populations are expanded in Apc-heterozygous mice compared with the control littermates; however, the HSCs have a reduced capacity to regenerate hematopoiesis in vivo in the absence of a single allele of Apc. Apc heterozygous myeloid progenitor cells display an increased frequency of apoptosis, and decreased in vitro colony-forming capacity, recapitulating several characteristic features of myeloid neoplasms with a -5/del(5q). Our results indicate that haploinsufficiency of Apc impairs hematopoiesis, and raise the possibility that loss of function of APC contributes to the development of myelodysplasia.


Subject(s)
Adenomatous Polyposis Coli Protein , Chromosome Deletion , Chromosomes, Mammalian , Erythropoiesis , Myeloid Progenitor Cells/metabolism , Anemia, Macrocytic/genetics , Anemia, Macrocytic/metabolism , Animals , Apoptosis/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Mice , Mice, Transgenic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism
16.
Semin Diagn Pathol ; 28(4): 304-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22195408

ABSTRACT

The classification and diagnostic criteria of the myeloproliferative neoplasms have changed significantly in the 2008 World Health Organization monograph on the classification of hematologic malignancies. Many of the changes arose from the findings that the different malignancies are associated with abnormal cell signaling because of translocations or mutations in genes for protein tryosine kinases involved in the normal growth and regulation of hematopoietic cells. These include ABL1, PDGFRA, PDGFRB, FGFR1, JAK2, MPL, and KIT. The new classification attempts to reflect the related molecular pathogenesis of the different entities and incorporates the identification of the molecular defects into the diagnostic criteria for some of the individual diseases. Issues concerning the new classification are discussed, and the new diagnostic criteria are reviewed and commented upon.


Subject(s)
Myeloproliferative Disorders/classification , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Protein-Tyrosine Kinases/genetics , Humans , Mutation
17.
Blood Cells Mol Dis ; 45(4): 317-23, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20817505

ABSTRACT

Although classically described as the driving oncogene in Burkitt lymphoma (BL), abnormalities of MYC have been recognized in other non-Hodgkin lymphomas as well. For example, MYC is overexpressed in approximately 10% of diffuse large B-cell lymphomas (DLBCL), conferring an adverse prognosis with chemoresistance and shortened survival; only approximately 30% of patients achieve long-term survival despite modern therapies. In contrast to BL, MYC aberrations in DLBCL are usually associated with multiple cytogenetic abnormalities and other genetic lesions, such as concurrent BCL2 translocations. Patients with so-called "double-hit" lymphomas have a worse outcome with few survivors beyond 6 months. It is unclear why MYC translocations are diagnostic in BL but prognostic in other lymphomas; different mechanisms underlying MYC abnormalities and a unique target set of genes may explain some of the variance. Furthermore, MYC possesses nontranscriptional functions other than transcriptional controls on genes regulating cell growth and may also influence the lymphoma microenvironment. Here we summarize current knowledge regarding MYC in lymphomas other than Burkitt lymphoma, with an emphasis on transcriptional, epigenetic, clinical, and microenvironmental consequences.


Subject(s)
Burkitt Lymphoma/diagnosis , Lymphoma/diagnosis , Proto-Oncogene Proteins c-myc/genetics , Burkitt Lymphoma/genetics , Epigenomics , Humans , Lymphoma/genetics , Translocation, Genetic
18.
Biol Blood Marrow Transplant ; 15(11): 1394-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19822298

ABSTRACT

We evaluated stem cell mobilization in 195 consecutive sibling donors who underwent a uniform mobilization regimen of granulocyte colony-stimulating factor (G-CSF) at 10 microg/kg/day divided into twice daily dosing. On day 5, peripheral blood (PB) CD34 cells/microL were measured immediately prior to peripheral blood stem cell (PBSC) apheresis. Failed mobilization was defined as <20 CD34 cells/microL on day 5. The median age was 52 years and 73 (37%) were 55 years or greater. Comorbid conditions by the Charlson Comorbidity Index (CCI) occurred in 13%, but only 3% had Karnofsky performance status (PS) <100%. Eight (4%) failed mobilization, defined as <20 CD34 cells/microL on day 5. Older age was associated with fewer CD34 cells/microL (P=.002). In addition, 6/73 (8.2%) older donors failed mobilization compared to 2/122 (1.6%) younger donors (P=.054). Comorbidity, sex, race, and donor weight did not influence mobilization. Although low PS was very uncommon, it was associated with reduced mobilization (P=.021), but not mobilization failure. A small fraction of older donors mobilize poorly, and this is not explained by standard measures of comorbidity or PS.


Subject(s)
Age Factors , Granulocyte Colony-Stimulating Factor/pharmacology , Health Status , Hematopoietic Stem Cell Mobilization/statistics & numerical data , Living Donors , Peripheral Blood Stem Cell Transplantation , Adolescent , Adult , Aged , Blood Cell Count , Body Weight , Comorbidity , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Humans , Male , Middle Aged , Recombinant Proteins , Siblings , Transplantation, Homologous , Young Adult
20.
Nat Commun ; 9(1): 1163, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29563491

ABSTRACT

The roles of RNA 5-methylcytosine (RNA:m5C) and RNA:m5C methyltransferases (RCMTs) in lineage-associated chromatin organization and drug response/resistance are unclear. Here we demonstrate that the RCMTs, namely NSUN3 and DNMT2, directly bind hnRNPK, a conserved RNA-binding protein. hnRNPK interacts with the lineage-determining transcription factors (TFs), GATA1 and SPI1/PU.1, and with CDK9/P-TEFb to recruit RNA-polymerase-II at nascent RNA, leading to formation of 5-Azacitidine (5-AZA)-sensitive chromatin structure. In contrast, NSUN1 binds BRD4 and RNA-polymerase-II to form an active chromatin structure that is insensitive to 5-AZA, but hypersensitive to the BRD4 inhibitor JQ1 and to the downregulation of NSUN1 by siRNAs. Both 5-AZA-resistant leukaemia cell lines and clinically 5-AZA-resistant myelodysplastic syndrome and acute myeloid leukaemia specimens have a significant increase in RNA:m5C and NSUN1-/BRD4-associated active chromatin. This study reveals novel RNA:m5C/RCMT-mediated chromatin structures that modulate 5-AZA response/resistance in leukaemia cells, and hence provides a new insight into treatment of leukaemia.


Subject(s)
Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , RNA, Neoplasm/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/chemistry , Chromatin/drug effects , Chromatin/metabolism , Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL