401
|
Patel C, Stenke L, Varma S, Lindberg ML, Björkholm M, Sjöberg J, Viktorsson K, Lewensohn R, Landgren O, Gottesman MM, Gillet JP. Multidrug resistance in relapsed acute myeloid leukemia: evidence of biological heterogeneity. Cancer 2013; 119:3076-83. [PMID: 23674237 DOI: 10.1002/cncr.28098] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/04/2013] [Indexed: 02/01/2023]
Abstract
BACKGROUND Studies of mechanisms mediating resistance to chemotherapy led to the discovery of the multidrug transporter ABCB1 (ATP-binding cassette, subfamily B, member 1), often expressed in leukemic cells of patients with acute myeloid leukemia (AML). Most clinical trials evaluating the strategy of inhibiting efflux-mediated chemotherapeutic resistance have been unsuccessful, clearly indicating the need for a better approach. METHODS This study investigated the clinical relevance of 380 genes whose expression has been shown to affect the response to chemotherapy, mostly through in vitro studies, in 11 paired samples obtained at AML diagnosis and at relapse. The expression profiling of these 380 genes was performed using TaqMan-based quantitative reverse-transcription polymerase chain reaction. Patients had a median age of 58 years at diagnosis, a median duration of complete remission of 284.5 days, and a median overall survival of 563 days. Cytogenetic abnormalities were detected at diagnosis in 4 patients, whereas 5 displayed a normal karyotype and 2 were not investigated. RESULTS Hierarchical clustering shows that samples taken at diagnosis and relapse clustered in pairs for 6 patients of the 11 studied, suggesting recurrence of the same leukemic blast, whereas for the other 5 patients, the data indicate their relapse blasts arose from different origins. A patient-by-patient analysis of the paired samples led to the striking observation that each had a unique gene signature representing different mechanisms of resistance. CONCLUSIONS The data underline the need for personalized molecular analysis to tailor treatment for patients with AML.
Collapse
Affiliation(s)
- Chirayu Patel
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
402
|
Roy S, Di Cello F, Kowalski J, Hristov AC, Tsai HL, Bhojwani D, Meyer JA, Carroll WL, Belton A, Resar LMS. HMGA1 overexpression correlates with relapse in childhood B-lineage acute lymphoblastic leukemia. Leuk Lymphoma 2013; 54:2565-7. [PMID: 23472968 DOI: 10.3109/10428194.2013.782610] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Sujayita Roy
- Hematology Division, The Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
403
|
Capalbo G, Mueller-Kuller T, Koschmieder S, Klein HU, Ottmann OG, Hoelzer D, Scheuring UJ. Characterization of ZC3H15 as a potential TRAF-2-interacting protein implicated in the NFκB pathway and overexpressed in AML. Int J Oncol 2013; 43:246-54. [PMID: 23624947 DOI: 10.3892/ijo.2013.1924] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 04/02/2013] [Indexed: 11/06/2022] Open
Abstract
The gene product of the zinc finger CCCH-type containing 15 (ZC3H15) gene, an immediate early erythropoietin response gene (synonymous: LEREPO4), was further characterized. ZC3H15 was expressed ubiquitously in all human tissues tested by northern blotting and showed mainly a diffuse cytoplasmic distribution by immune fluorescence microscopy and western blotting of subcellular protein fractions. The expression of ZC3H15 was downregulated effectively in HeLa cells to ≤13% of the control by transfection of specific small interfering RNA (siRNA). Subsequent Affymetrix microarray analysis revealed 202 differentially expressed genes including 114 induced (≥3-fold) genes and 88 suppressed (≤0.3-fold) genes. The gene ontology (GO) categories containing an over-representation of differentially expressed genes comprised cell growth, transcription, cell adhesion, regulation of NF-κB, regulation of MAPK, cell cycle arrest and immune response. ZC3H15 interacted with the signaling adapter protein tumor necrosis factor receptor associated factor 2 (TRAF-2) as shown by co-immunoprecipitation. ZC3H15 expression was found to be significantly increased in acute myeloid leukemia (AML) samples compared to MDS, CML, ALL and normal bone marrow samples using the Leukemia Gene Atlas (LGA) database. Based on these data, it is hypothesized that ZC3H15 may interact with TRAF-2 functionally within the NF-κB pathway, and may be explored as a potential target in AML.
Collapse
Affiliation(s)
- Gianni Capalbo
- Department of Hematology/Oncology and Infectious Diseases, J.W. Goethe-University Hospital, 60595 Frankfurt/Main, Germany
| | | | | | | | | | | | | |
Collapse
|
404
|
Targeting oncogene expression to endothelial cells induces proliferation of the myelo-erythroid lineage by repressing the notch pathway. Leukemia 2013; 27:2229-41. [DOI: 10.1038/leu.2013.132] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 11/08/2022]
|
405
|
Storry JR, Jöud M, Christophersen MK, Thuresson B, Åkerström B, Sojka BN, Nilsson B, Olsson ML. Homozygosity for a null allele of SMIM1 defines the Vel-negative blood group phenotype. Nat Genet 2013; 45:537-41. [PMID: 23563606 DOI: 10.1038/ng.2600] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/06/2013] [Indexed: 11/09/2022]
Abstract
The Vel antigen is present on red blood cells (RBCs) from all humans except rare Vel-negative individuals who can form antibodies to Vel in response to transfusion or pregnancy. These antibodies may cause severe hemolytic reactions in blood recipients. We combined SNP profiling and transcriptional network modeling to link the Vel-negative phenotype to SMIM1, located in a 97-kb haplotype block on chromosome 1p36. This gene encodes a previously undiscovered, evolutionarily conserved transmembrane protein expressed on RBCs. Notably, 35 of 35 Vel-negative individuals were homozygous for a frameshift deletion of 17 bp in exon 3. Functional studies using antibodies raised against SMIM1 peptides confirmed a null phenotype in RBC membranes, and SMIM1 overexpression induced Vel expression. Genotype screening estimated that ~1 of 17 Swedish blood donors is a heterozygous deletion carrier and ~1 of 1,200 is a homozygous deletion knockout and enabled identification of Vel-negative donors. Our results establish SMIM1 as a new erythroid gene and Vel as a new blood group system.
Collapse
Affiliation(s)
- Jill R Storry
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
406
|
Abstract
There is growing research interest in the mammalian Tribbles (Trib) family of serine/threonine pseudokinases and their oncogenic association with acute leukemias. This review is to understand the role of Trib genes in hematopoietic malignancies and their potential as targets for novel therapeutic strategies in acute myeloid leukemia and acute lymphoblastic leukemia. We discuss the role of Tribs as central signaling mediators in different subtypes of acute leukemia and propose that inhibition of dysregulated Trib signaling may be therapeutically beneficial.
Collapse
|
407
|
Dickson GJ, Liberante FG, Kettyle LM, O'Hagan KA, Finnegan DPJ, Bullinger L, Geerts D, McMullin MF, Lappin TRJ, Mills KI, Thompson A. HOXA/PBX3 knockdown impairs growth and sensitizes cytogenetically normal acute myeloid leukemia cells to chemotherapy. Haematologica 2013; 98:1216-25. [PMID: 23539541 DOI: 10.3324/haematol.2012.079012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The cytogenetically normal subtype of acute myeloid leukemia is associated with an intermediate risk which complicates therapeutic options. Lower overall HOX/TALE expression appears to correlate with more favorable prognosis/better response to treatment in some leukemias and solid cancer. The functional significance of the associated gene expression and response to chemotherapy is not known. Three independent microarray datasets obtained from large cohorts of patients along with quantitative polymerase chain reaction validation were used to identify a four-gene HOXA/TALE signature capable of prognostic stratification. Biochemical analysis was used to identify interactions between the four encoded proteins and targeted knockdown used to examine the functional importance of sustained expression of the signature in leukemia maintenance and response to chemotherapy. An 11 HOXA/TALE code identified in an intermediate-risk group of patients (n=315) compared to a group with a favorable risk (n=105) was reduced to a four-gene signature of HOXA6, HOXA9, PBX3 and MEIS1 by iterative analysis of independent platforms. This signature maintained the favorable/intermediate risk partition and where applicable, correlated with overall survival in cytogenetically normal acute myeloid leukemia. We further showed that cell growth and function are dependent on maintained levels of these core genes and that direct targeting of HOXA/PBX3 sensitizes cytogenetically normal acute myeloid leukemia cells to standard chemotherapy. Together the data support a key role for HOXA/TALE in cytogenetically normal acute myeloid leukemia and demonstrate that targeting of clinically significant HOXA/PBX3 elements may provide therapeutic benefit to patients with this subtype of leukemia.
Collapse
Affiliation(s)
- Glenda J Dickson
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Northern Ireland, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
408
|
Coskun E, Neumann M, Schlee C, Liebertz F, Heesch S, Goekbuget N, Hoelzer D, Baldus CD. MicroRNA profiling reveals aberrant microRNA expression in adult ETP-ALL and functional studies implicate a role for miR-222 in acute leukemia. Leuk Res 2013; 37:647-56. [PMID: 23522449 DOI: 10.1016/j.leukres.2013.02.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 01/14/2013] [Accepted: 02/20/2013] [Indexed: 12/12/2022]
Abstract
Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) has been identified as high-risk subgroup in acute T-cell lymphoblastic leukemia (T-ALL). To investigate the immature and myeloid nature of ETP-ALL we examined global microRNA (miRNA) expression in adult ETP-ALL. miRNA profiling of ETP-ALL (n=8), non-ETP T-ALL (n=6), and healthy controls was performed and results were validated in independent cohorts of 66 ETP-ALL and 111 non-ETP T-ALL using real-time RT-PCR. Furthermore, in vitro studies were performed on deregulated miRNAs in acute leukemia. We identified miR-221 and miR-222 as the most upregulated and six miRNAs (miR-151-3p, miR-19a, miR-20b, miR-342-3p, miR-363, and miR-576-3p) as downregulated in ETP-ALL compared to non-ETP T-ALL. In the validation cohorts, miR-221 and miR-222 were significantly upregulated in ETP-ALL, and miR-363 and miR-19a were downregulated in ETP-ALL. ETS1, downregulated in ETP-ALL, was identified as direct target of miR-222. In our in vitro studies miR-222 significantly inhibited proliferation, and caused cell cycle arrest and apoptosis in leukemic cells. In conclusion, our study revealed aberrant miRNA expression in ETP-ALL, with miR-221 and miR-222 as the most overexpressed miRNAs and implied a functional role for miR-222 in leukemic cells. Importantly, miR-222 may impact leukemogenesis by altering expression of the proto-oncogene ETS1 in acute leukemia.
Collapse
Affiliation(s)
- Ebru Coskun
- Hematology and Oncology, Charité, University Hospital Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
409
|
Development of gene expression-based risk score in cytogenetically normal acute myeloid leukemia patients. Oncotarget 2013; 3:824-32. [PMID: 22910040 PMCID: PMC3478459 DOI: 10.18632/oncotarget.571] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Patients with normal karyotype represent the single largest cytogenetic group of acute myeloid leukemia (AML), with highly heterogeneous clinical and molecular characteristics. In this study, we sought to determine new prognostic biomarkers in cytogenetically normal (CN)-AML patients. A gene expression (GE)-based risk score was built, summing up the prognostic value of 22 genes whose expression is associated with a bad prognosis in a training cohort of 163 patients. GE-based risk score allowed identifying a high-risk group of patients (53.4%) in two independent cohorts of CN-AML patients. GE-based risk score and EVI1 gene expression remained independent prognostic factors using multivariate Cox analyses. Combining GE-based risk score with EVI1 gene expression allowed the identification of three clinically different groups of patients in two independent cohorts of CN-AML patients. Thus, GE-based risk score is powerful to predict clinical outcome for CN-AML patients and may provide potential therapeutic advances.
Collapse
|
410
|
Friedman DR, Lucas JE, Weinberg JB. Clinical and biological relevance of genomic heterogeneity in chronic lymphocytic leukemia. PLoS One 2013; 8:e57356. [PMID: 23468975 PMCID: PMC3585365 DOI: 10.1371/journal.pone.0057356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/21/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) is typically regarded as an indolent B-cell malignancy. However, there is wide variability with regards to need for therapy, time to progressive disease, and treatment response. This clinical variability is due, in part, to biological heterogeneity between individual patients' leukemias. While much has been learned about this biological variation using genomic approaches, it is unclear whether such efforts have sufficiently evaluated biological and clinical heterogeneity in CLL. METHODS To study the extent of genomic variability in CLL and the biological and clinical attributes of genomic classification in CLL, we evaluated 893 unique CLL samples from fifteen publicly available gene expression profiling datasets. We used unsupervised approaches to divide the data into subgroups, evaluated the biological pathways and genetic aberrations that were associated with the subgroups, and compared prognostic and clinical outcome data between the subgroups. RESULTS Using an unsupervised approach, we determined that approximately 600 CLL samples are needed to define the spectrum of diversity in CLL genomic expression. We identified seven genomically-defined CLL subgroups that have distinct biological properties, are associated with specific chromosomal deletions and amplifications, and have marked differences in molecular prognostic markers and clinical outcomes. CONCLUSIONS Our results indicate that investigations focusing on small numbers of patient samples likely provide a biased outlook on CLL biology. These findings may have important implications in identifying patients who should be treated with specific targeted therapies, which could have efficacy against CLL cells that rely on specific biological pathways.
Collapse
Affiliation(s)
- Daphne R Friedman
- Department of Medicine, Duke University, Durham, North Carolina, USA.
| | | | | |
Collapse
|
411
|
Yang J, Schiffer CA. Genetic biomarkers in acute myeloid leukemia: will the promise of improving treatment outcomes be realized? Expert Rev Hematol 2013; 5:395-407. [PMID: 22992234 DOI: 10.1586/ehm.12.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recent progress in the molecular genetics of acute myeloid leukemia (AML) has shown this disease to be more heterogeneous than previously realized. Recurrent cytogenetic and mutational changes in leukemic blasts have been confirmed to have high prognostic significance. High-throughput techniques to analyze the AML genome in greater depth have revealed novel mutations with putative roles in leukemogenesis. The use of prognostic biomarkers has allowed for a more detailed categorization of AML based on risk. Despite this tremendous progress, the understanding of the mechanisms by which these changes influence leukemia growth and response to treatment is still limited, which in turn has hindered the development of rationally targeted therapies for AML. The integration of clinical, cytogenetic and molecular data will be essential to translate the current research momentum into better outcomes for patients with AML.
Collapse
Affiliation(s)
- Jay Yang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| | | |
Collapse
|
412
|
Ono M, Tanaka RJ, Kano M, Sugiman T. Visualising the cross-level relationships between pathological and physiological processes and gene expression: analyses of haematological diseases. PLoS One 2013; 8:e53544. [PMID: 23301083 PMCID: PMC3534650 DOI: 10.1371/journal.pone.0053544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/03/2012] [Indexed: 11/18/2022] Open
Abstract
The understanding of pathological processes is based on the comparison between physiological and pathological conditions, and transcriptomic analysis has been extensively applied to various diseases for this purpose. However, the way in which the transcriptomic data of pathological cells relate to the transcriptomes of normal cellular counterparts has not been fully explored, and may provide new and unbiased insights into the mechanisms of these diseases. To achieve this, it is necessary to develop a method to simultaneously analyse components across different levels, namely genes, normal cells, and diseases. Here we propose a multidimensional method that visualises the cross-level relationships between these components at three different levels based on transcriptomic data of physiological and pathological processes, by adapting Canonical Correspondence Analysis, which was developed in ecology and sociology, to microarray data (CCA on Microarray data, CCAM). Using CCAM, we have analysed transcriptomes of haematological disorders and those of normal haematopoietic cell differentiation. First, by analysing leukaemia data, CCAM successfully visualised known relationships between leukaemia subtypes and cellular differentiation, and their characteristic genes, which confirmed the relevance of CCAM. Next, by analysing transcriptomes of myelodysplastic syndromes (MDS), we have shown that CCAM was effective in both generating and testing hypotheses. CCAM showed that among MDS patients, high-risk patients had transcriptomes that were more similar to those of both haematopoietic stem cells (HSC) and megakaryocyte-erythroid progenitors (MEP) than low-risk patients, and provided a prognostic model. Collectively, CCAM reveals hidden relationships between pathological and physiological processes and gene expression, providing meaningful clinical insights into haematological diseases, and these could not be revealed by other univariate and multivariate methods. Furthermore, CCAM was effective in identifying candidate genes that are correlated with cellular phenotypes of interest. We expect that CCAM will benefit a wide range of medical fields.
Collapse
Affiliation(s)
- Masahiro Ono
- Immunobiology Unit, Institute of Child Health, University College London, London, United Kingdom.
| | | | | | | |
Collapse
|
413
|
Qazi S, Ma H, Uckun FM. Absence of Genomic Ikaros/IKZF1 Deletions in Pediatric B-Precursor Acute Lymphoblastic Leukemia. ACTA ACUST UNITED AC 2013; 3:72-82. [PMID: 24478816 DOI: 10.5376/ijmms.2013.03.0009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Here we report the results of gene expression analyses using multiple probesets aimed at determining the incidence of Ikaros/IKZF1 deletions in pediatric B-precursor acute lymphoblastic leukemia (BPL). Primary leukemia cells from 122 Philadelphia chromosome (Ph)+ BPL patients and 237 Ph- BPL patients as well as normal hematopoietic cells from 74 normal non-leukemic bone marrow specimens were organized according to expression levels of IKZF1 transcripts utilizing two-way hierarchical clustering technique to identify specimens with low IKZF1 expression for the 10 probesets interrogating Exons 1 through 4 and Exon 8. Our analysis demonstrated no changes in expression that would be expected from homozygous or heterozygous deletions of IKZF1 in primary leukemic cells. Similar results were obtained in gene expression analysis of primary leukemic cells from 20 Ph+ positive and 155 Ph- BPL patients in a validation dataset. Taken together, our gene expression analyses in 534 pediatric BPL cases, including 142 cases with Ph+ BPL, contradict previous reports that were based on SNP array data and suggested that Ph+ pediatric BPL is characterized by a high frequency of homozygous or heterozygous IKZF1 deletions. Further, exon-specific genomic PCR analysis of primary leukemia cells from 21 high-risk pediatric BPL patients and 11 standard-risk pediatric BPL patients, and 8 patients with infant BPL did not show any evidence for homozygous IKZF1 locus deletions. Nor was there any evidence for homozygous or heterozygous intragenic IKZF1 deletions.
Collapse
Affiliation(s)
- Sanjive Qazi
- Systems Immunobiology Laboratory, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027 ; Department of Biology and Bioinformatics Program, Gustavus Adolphus College, 800 W College Avenue, St. Peter, MN 56082
| | - Hong Ma
- Systems Immunobiology Laboratory, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027
| | - Fatih M Uckun
- Systems Immunobiology Laboratory, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027 ; Department of Biology and Bioinformatics Program, Gustavus Adolphus College, 800 W College Avenue, St. Peter, MN 56082 ; Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA
| |
Collapse
|
414
|
Gatt ME, Ben-Yehuda D, Izraeli S. Lymphoid leukemias. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00092-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
415
|
Li K, Yang M, Sablok G, Fan J, Zhou F. Screening features to improve the class prediction of acute myeloid leukemia and myelodysplastic syndrome. Gene 2013; 512:348-54. [DOI: 10.1016/j.gene.2012.09.123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 09/29/2012] [Indexed: 02/06/2023]
|
416
|
Järvstråt L, Johansson M, Gullberg U, Nilsson B. Ultranet: efficient solver for the sparse inverse covariance selection problem in gene network modeling. Bioinformatics 2012; 29:511-2. [DOI: 10.1093/bioinformatics/bts717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
417
|
Ofran Y, Rowe JM. Genetic profiling in acute myeloid leukaemia ─ where are we and what is its role in patient management. Br J Haematol 2012; 160:303-20. [DOI: 10.1111/bjh.12135] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
418
|
Abstract
Myelodysplastic syndromes (MDS) are malignant clonal disorders of haematopoietic stem cells and their microenvironment, affecting older individuals (median age ∼70 years). Unique features that are associated with MDS - but which are not necessarily present in every patient with MDS - include excessive apoptosis in maturing clonal cells, a pro-inflammatory bone marrow microenvironment, specific chromosomal abnormalities, abnormal ribosomal protein biogenesis, the presence of uniparental disomy, and mutations affecting genes involved in proliferation, methylation and epigenetic modifications. Although emerging insights establish an association between molecular abnormalities and the phenotypic heterogeneity of MDS, their origin and progression remain enigmatic.
Collapse
Affiliation(s)
- Azra Raza
- Myelodysplastic Syndromes Center, Columbia University Medical Center, Milstein Hospital Building, 6N-435, 177 Fort Washington Avenue, New York, New York 10032, USA.
| | | |
Collapse
|
419
|
Jamil K, Jayaraman A, Rao R, Raju S. In silico evidence of signaling pathways of notch mediated networks in leukemia. Comput Struct Biotechnol J 2012; 1:e201207005. [PMID: 24688641 PMCID: PMC3962152 DOI: 10.5936/csbj.201207005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 11/04/2012] [Accepted: 11/07/2012] [Indexed: 11/22/2022] Open
Abstract
Notch signaling plays a critical role in cell fate determination and maintenance of progenitors in many developmental systems. Notch receptors have been shown to be expressed on hematopoietic progenitor cells as well as to various degrees in peripheral blood T and B lymphocytes, monocytes, and neutrophils. Our aim was to understand the protein interaction network, using Notch1 protein name as query in STRING database and we generated a model to assess the significance of Notch1 associated proteins in Acute Lymphoblastic Leukemia (ALL). We further analyzed the expression levels of the genes encoding hub proteins, using Oncomine database, to determine their significance in leukemogenesis. Of the forty two hub genes, we observed that sixteen genes were underexpressed and eleven genes were overexpressed in T-cell Acute Lymphoblastic samples in comparison to their expression levels in normal cells. Of these, we found three novel genes which have not been reported earlier- KAT2B, PSEN1 (underexpressed) and CDH2 (overexpressed).These three identified genes may provide new insights into the abnormal hematopoietic process observed in Leukemia as these genes are involved in Notch signaling and cell adhesion processes. It is evident that experimental validation of the protein interactors in leukemic cells could help in the identification of new diagnostic markers for leukemia.
Collapse
Affiliation(s)
- Kaiser Jamil
- Centre for Biotechnology and Bioinformatics, School of Life sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Budha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India
| | - Archana Jayaraman
- Centre for Biotechnology and Bioinformatics, School of Life sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Budha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India
| | - Raghunatha Rao
- Oncology Department, Nizams Institute of Medical Sciences ( NIMS), Panjagutta, Hyderabad 500082, Andhra Pradesh, India
| | - Suryanarayana Raju
- Oncology Department, Nizams Institute of Medical Sciences ( NIMS), Panjagutta, Hyderabad 500082, Andhra Pradesh, India
| |
Collapse
|
420
|
Coletta A, Molter C, Duqué R, Steenhoff D, Taminau J, de Schaetzen V, Meganck S, Lazar C, Venet D, Detours V, Nowé A, Bersini H, Weiss Solís DY. InSilico DB genomic datasets hub: an efficient starting point for analyzing genome-wide studies in GenePattern, Integrative Genomics Viewer, and R/Bioconductor. Genome Biol 2012; 13:R104. [PMID: 23158523 PMCID: PMC3580496 DOI: 10.1186/gb-2012-13-11-r104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 11/18/2012] [Indexed: 12/18/2022] Open
Abstract
Genomics datasets are increasingly useful for gaining biomedical insights, with adoption in the clinic underway. However, multiple hurdles related to data management stand in the way of their efficient large-scale utilization. The solution proposed is a web-based data storage hub. Having clear focus, flexibility and adaptability, InSilico DB seamlessly connects genomics dataset repositories to state-of-the-art and free GUI and command-line data analysis tools. The InSilico DB platform is a powerful collaborative environment, with advanced capabilities for biocuration, dataset sharing, and dataset subsetting and combination. InSilico DB is available from https://insilicodb.org.
Collapse
|
421
|
Starmans MHW, Pintilie M, John T, Der SD, Shepherd FA, Jurisica I, Lambin P, Tsao MS, Boutros PC. Exploiting the noise: improving biomarkers with ensembles of data analysis methodologies. Genome Med 2012; 4:84. [PMID: 23146350 PMCID: PMC3580418 DOI: 10.1186/gm385] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/04/2012] [Accepted: 11/12/2012] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The advent of personalized medicine requires robust, reproducible biomarkers that indicate which treatment will maximize therapeutic benefit while minimizing side effects and costs. Numerous molecular signatures have been developed over the past decade to fill this need, but their validation and up-take into clinical settings has been poor. Here, we investigate the technical reasons underlying reported failures in biomarker validation for non-small cell lung cancer (NSCLC). METHODS We evaluated two published prognostic multi-gene biomarkers for NSCLC in an independent 442-patient dataset. We then systematically assessed how technical factors influenced validation success. RESULTS Both biomarkers validated successfully (biomarker #1: hazard ratio (HR) 1.63, 95% confidence interval (CI) 1.21 to 2.19, P = 0.001; biomarker #2: HR 1.42, 95% CI 1.03 to 1.96, P = 0.030). Further, despite being underpowered for stage-specific analyses, both biomarkers successfully stratified stage II patients and biomarker #1 also stratified stage IB patients. We then systematically evaluated reasons for reported validation failures and find they can be directly attributed to technical challenges in data analysis. By examining 24 separate pre-processing techniques we show that minor alterations in pre-processing can change a successful prognostic biomarker (HR 1.85, 95% CI 1.37 to 2.50, P < 0.001) into one indistinguishable from random chance (HR 1.15, 95% CI 0.86 to 1.54, P = 0.348). Finally, we develop a new method, based on ensembles of analysis methodologies, to exploit this technical variability to improve biomarker robustness and to provide an independent confidence metric. CONCLUSIONS Biomarkers comprise a fundamental component of personalized medicine. We first validated two NSCLC prognostic biomarkers in an independent patient cohort. Power analyses demonstrate that even this large, 442-patient cohort is under-powered for stage-specific analyses. We then use these results to discover an unexpected sensitivity of validation to subtle data analysis decisions. Finally, we develop a novel algorithmic approach to exploit this sensitivity to improve biomarker robustness.
Collapse
Affiliation(s)
- Maud HW Starmans
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Melania Pintilie
- Ontario Cancer Institute and the Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Thomas John
- Ludwig Institute for Cancer Research, Austin Health, Melbourne, Australia
| | - Sandy D Der
- Ontario Cancer Institute and the Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Frances A Shepherd
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Igor Jurisica
- Ontario Cancer Institute and the Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 2M9, Canada
- Techna Institute, University Health Network, Toronto, ON, M5G 2M9, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Philippe Lambin
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ming-Sound Tsao
- Ontario Cancer Institute and the Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Paul C Boutros
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 2M9, Canada
| |
Collapse
|
422
|
Fernandez P, Solenthaler M, Spertini O, Quarroz S, Rovo A, Lovey PY, Leoncini L, Ruault-Jungblut S, D’Asaro M, Schaad O, Docquier M, Descombes P, Matthes T. Using digital RNA counting and flow cytometry to compare mRNA with protein expression in acute leukemias. PLoS One 2012; 7:e49010. [PMID: 23152841 PMCID: PMC3494663 DOI: 10.1371/journal.pone.0049010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 10/03/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The diagnosis of malignant hematologic diseases has become increasingly complex during the last decade. It is based on the interpretation of results from different laboratory analyses, which range from microscopy to gene expression profiling. Recently, a method for the analysis of RNA phenotypes has been developed, the nCounter technology (Nanostring® Technologies), which allows for simultaneous quantification of hundreds of RNA molecules in biological samples. We evaluated this technique in a Swiss multi-center study on eighty-six samples from acute leukemia patients. METHODS mRNA and protein profiles were established for normal peripheral blood and bone marrow samples. Signal intensities of the various tested antigens with surface expression were similar to those found in previously performed Affymetrix microarray analyses. Acute leukemia samples were analyzed for a set of twenty-two validated antigens and the Pearson Correlation Coefficient for nCounter and flow cytometry results was calculated. RESULTS Highly significant values between 0.40 and 0.97 were found for the twenty-two antigens tested. A second correlation analysis performed on a per sample basis resulted in concordant results between flow cytometry and nCounter in 44-100% of the antigens tested (mean = 76%), depending on the number of blasts present in a sample, the homogeneity of the blast population, and the type of leukemia (AML or ALL). CONCLUSIONS The nCounter technology allows for fast and easy depiction of a mRNA profile from hematologic samples. This technology has the potential to become a valuable tool for the diagnosis of acute leukemias, in addition to multi-color flow cytometry.
Collapse
Affiliation(s)
| | - Max Solenthaler
- University Clinic for Hematology and Central Hematology Laboratory, Inselspital Bern and University of Bern, Bern, Switzerland
| | - Olivier Spertini
- Hematology Service, CHUV, University Hospital Lausanne, Lausanne, Switzerland
| | - Stephane Quarroz
- Hematology Service, CHUV, University Hospital Lausanne, Lausanne, Switzerland
| | - Alicia Rovo
- Hematology Department, University Hospital Basel, Basel, Switzerland
| | - Pierre-Yves Lovey
- Hematology Service, Institut Central des Hôpitaux Valaisans, Sion, Switzerland
| | - Leda Leoncini
- Hematology Service, Istituto Oncologico della Svizzera Italiana, Bellinzona, Switzerland
| | | | - Mathilde D’Asaro
- Hematology Service, University Hospital Geneva, Geneva, Switzerland
| | - Olivier Schaad
- Genomics Platform, CMU, University Medical Center, Geneva, Switzerland
| | - Mylène Docquier
- Genomics Platform, CMU, University Medical Center, Geneva, Switzerland
| | - Patrick Descombes
- Genomics Platform, CMU, University Medical Center, Geneva, Switzerland
| | - Thomas Matthes
- Hematology Service, University Hospital Geneva, Geneva, Switzerland
- * E-mail:
| | | |
Collapse
|
423
|
Bagger FO, Rapin N, Theilgaard-Mönch K, Kaczkowski B, Thoren LA, Jendholm J, Winther O, Porse BT. HemaExplorer: a database of mRNA expression profiles in normal and malignant haematopoiesis. Nucleic Acids Res 2012; 41:D1034-9. [PMID: 23143109 PMCID: PMC3531225 DOI: 10.1093/nar/gks1021] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The HemaExplorer (http://servers.binf.ku.dk/hemaexplorer) is a curated database of processed mRNA Gene expression profiles (GEPs) that provides an easy display of gene expression in haematopoietic cells. HemaExplorer contains GEPs derived from mouse/human haematopoietic stem and progenitor cells as well as from more differentiated cell types. Moreover, data from distinct subtypes of human acute myeloid leukemia is included in the database allowing researchers to directly compare gene expression of leukemic cells with those of their closest normal counterpart. Normalization and batch correction lead to full integrity of the data in the database. The HemaExplorer has comprehensive visualization interface that can make it useful as a daily tool for biologists and cancer researchers to assess the expression patterns of genes encountered in research or literature. HemaExplorer is relevant for all research within the fields of leukemia, immunology, cell differentiation and the biology of the haematopoietic system.
Collapse
Affiliation(s)
- Frederik Otzen Bagger
- Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, DK2200 Denmark
| | | | | | | | | | | | | | | |
Collapse
|
424
|
Taslaman L, Nilsson B. A framework for regularized non-negative matrix factorization, with application to the analysis of gene expression data. PLoS One 2012; 7:e46331. [PMID: 23133590 PMCID: PMC3487913 DOI: 10.1371/journal.pone.0046331] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 08/31/2012] [Indexed: 12/19/2022] Open
Abstract
Non-negative matrix factorization (NMF) condenses high-dimensional data into lower-dimensional models subject to the requirement that data can only be added, never subtracted. However, the NMF problem does not have a unique solution, creating a need for additional constraints (regularization constraints) to promote informative solutions. Regularized NMF problems are more complicated than conventional NMF problems, creating a need for computational methods that incorporate the extra constraints in a reliable way. We developed novel methods for regularized NMF based on block-coordinate descent with proximal point modification and a fast optimization procedure over the alpha simplex. Our framework has important advantages in that it (a) accommodates for a wide range of regularization terms, including sparsity-inducing terms like the penalty, (b) guarantees that the solutions satisfy necessary conditions for optimality, ensuring that the results have well-defined numerical meaning, (c) allows the scale of the solution to be controlled exactly, and (d) is computationally efficient. We illustrate the use of our approach on in the context of gene expression microarray data analysis. The improvements described remedy key limitations of previous proposals, strengthen the theoretical basis of regularized NMF, and facilitate the use of regularized NMF in applications.
Collapse
Affiliation(s)
- Leo Taslaman
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Björn Nilsson
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
- Broad Institute, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
425
|
Lee TY, Ezelle HJ, Venkataraman T, Lapidus RG, Scheibner KA, Hassel BA. Regulation of human RNase-L by the miR-29 family reveals a novel oncogenic role in chronic myelogenous leukemia. J Interferon Cytokine Res 2012; 33:34-42. [PMID: 23113544 DOI: 10.1089/jir.2012.0062] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The endoribonuclease RNase-L is the terminal component of an interferon-regulated RNA decay pathway known as the 2'-5'-oligoadenylate (2-5A) system, whose established functions include antimicrobial and tumor suppressive activities. RNase-L activity requires binding of the small molecule 2-5A, leading to RNase-L dimerization and cleavage of single-stranded RNA. RNase-L expression is controlled post-transcriptionally by its 3'-untranslated region (3' UTR), which exerts a strong negative effect on RNase-L levels. MicroRNAs (miRNAs) are a class of small noncoding RNAs that repress expression of target genes by binding to regions of complementarity often in the 3' UTR. The miR-29 family acts as a tumor suppressor in several cancers, including acute and chronic myelogenous leukemia (CML), and has many oncogenic targets. We report that the miR-29 family represses RNase-L protein expression across several cell types. Using a luciferase reporter, we showed that miR-29 acts via 4 target sites within the RNASEL 3' UTR. Mutation of all sites is required for abrogation of miR-29 repression. In light of the reported tumor suppressive role of miR-29 in K562 CML cells and miR-29 repression of RNase-L in these cells, we generated K562 cells with stable RNase-L knockdown and demonstrated that loss of RNase-L inhibits proliferation in vitro as well as tumor growth in a xenograft model. Our findings identify a previously unknown miRNA regulator of RNase-L expression and support a novel oncogenic role for RNase-L in CML and potentially other hematopoietic malignancies.
Collapse
Affiliation(s)
- Teresa Y Lee
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | |
Collapse
|
426
|
Lin S, Tian L, Shen H, Gu Y, Li JL, Chen Z, Sun X, You MJ, Wu L. DDX5 is a positive regulator of oncogenic NOTCH1 signaling in T cell acute lymphoblastic leukemia. Oncogene 2012; 32:4845-53. [PMID: 23108395 DOI: 10.1038/onc.2012.482] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 08/31/2012] [Accepted: 09/02/2012] [Indexed: 02/02/2023]
Abstract
Notch signaling is a highly conserved cell-cell communication pathway regulating normal development and tissue homeostasis. Aberrant Notch signaling represents an important oncogenic mechanism for T cell acute lymphoblastic leukemia (T-ALL), an aggressive subset of the most common malignant childhood cancer ALL. Therefore, understanding the molecular regulation of Notch signaling is critical to identify new approaches to block aberrant Notch oncogenic activity. The family of three MAML transcriptional coactivators is crucial for Notch signaling activation. The prototypic member MAML1 is the major coactivator that regulates Notch oncogenic activities in leukemic cells. However, the molecular basis underlying MAML1 coactivator function that contributes to Notch signaling remains unclear. In this study, we performed proteomic studies and identified DDX5, an ATP-dependent DEAD-box RNA helicase, as a component of the MAML1 protein complex. DDX5 interacts with MAML1 in vitro and in vivo, and is associated with the endogenous NOTCH1 transcription activation complex in human T-ALL leukemic cells. Lentivirus-mediated short-hairpin RNA knock-down of DDX5 resulted in decreased expression of Notch target genes, reduced cell proliferation and increased apoptosis in cultured human leukemic cells with constitutive activation of Notch signaling. Also, DDX5 depletion inhibited the growth of human leukemia xenograft in nude mice. Moreover, DDX5 is highly expressed in primary human T-ALL leukemic cells based on the analyses of Oncomine and GEO databases, and Immunohistochemical staining. Our overall findings revealed a critical role of DDX5 in promoting efficient Notch-mediated transcription in leukemic cells, suggesting that DDX5 might be critical for NOTCH1-mediated T-ALL pathogenesis and thus is a potential new target for modulating the Notch signaling in leukemia.
Collapse
Affiliation(s)
- S Lin
- Department of Molecular Genetics and Microbiology, Shands Cancer Center, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
427
|
Pigazzi M, Manara E, Bresolin S, Tregnago C, Beghin A, Baron E, Giarin E, Cho EC, Masetti R, Rao DS, Sakamoto KM, Basso G. MicroRNA-34b promoter hypermethylation induces CREB overexpression and contributes to myeloid transformation. Haematologica 2012; 98:602-10. [PMID: 23100280 DOI: 10.3324/haematol.2012.070664] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNA-34b down-regulation in acute myeloid leukemia was previously shown to induce CREB overexpression, thereby causing leukemia proliferation in vitro and in vivo. The role of microRNA-34b and CREB in patients with myeloid malignancies has never been evaluated. We examined microRNA-34b expression and the methylation status of its promoter in cells from patients diagnosed with myeloid malignancies. We used gene expression profiling to identify signatures of myeloid transformation. We established that microRNA-34b has suppressor ability and that CREB has oncogenic potential in primary bone marrow cell cultures and in vivo. MicroRNA-34b was found to be up-regulated in pediatric patients with juvenile myelomonocytic leukemia (n=17) and myelodysplastic syndromes (n=28), but was down-regulated in acute myeloid leukemia patients at diagnosis (n=112). Our results showed that hypermethylation of the microRNA-34b promoter occurred in 66% of cases of acute myeloid leukemia explaining the low microRNA-34b levels and CREB overexpression, whereas preleukemic myelodysplastic syndromes and juvenile myelomonocytic leukemia were not associated with hypermethylation or CREB overexpression. In paired samples taken from the same patients when they had myelodysplastic syndrome and again during the subsequent acute myeloid leukemia, we confirmed microRNA-34b promoter hypermethylation at leukemia onset, with 103 CREB target genes differentially expressed between the two disease stages. This subset of CREB targets was confirmed to associate with high-risk myelodysplastic syndromes in a separate cohort of patients (n=20). Seventy-eight of these 103 CREB targets were also differentially expressed between healthy samples (n=11) and de novo acute myeloid leukemia (n=72). Further, low microRNA-34b and high CREB expression levels induced aberrant myelopoiesis through CREB-dependent pathways in vitro and in vivo. In conclusion, we suggest that microRNA-34b controls CREB expression and contributes to myeloid transformation from both healthy bone marrow and myelodysplastic syndromes. We identified a subset of CREB target genes that represents a novel transcriptional network that may control myeloid transformation.
Collapse
Affiliation(s)
- Martina Pigazzi
- Women and Child Health Department, Hematology-Oncology Laboratory, University of Padova, Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
428
|
Abstract
The family with sequence similarity 83, member D (Fam83D) encodes a mitotic spindle-associated protein. Its knockdown results in shorter spindles that fail to organize a correct metaphase plate. In this study, we demonstrated that Fam83D is coexpressed with well-known mitotic genes. Pathway analysis results also showed that cell cycle- and mitosis-related pathways are enriched with Fam83D-coexpressed genes. Furthermore, Fam83D is differentially expressed in various types of cancers. The results presented in this study suggest that Fam83D may be an important molecule for mitotic progression and equal segregation of chromosomes. Since the molecules that are involved in these mechanisms are crucial for mitosis as well as carcinogenesis, Fam83D should be considered as a novel regulator of mitosis and a putative carcinogenesis-related gene.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Biology, Faculty of Science, Harran University, Osmanbey Campus, Sanliurfa, Turkey
| |
Collapse
|
429
|
del Rey M, O'Hagan K, Dellett M, Aibar S, Colyer HAA, Alonso ME, Díez-Campelo M, Armstrong RN, Sharpe DJ, Gutiérrez NC, García JL, De Las Rivas J, Mills KI, Hernández-Rivas JM. Genome-wide profiling of methylation identifies novel targets with aberrant hypermethylation and reduced expression in low-risk myelodysplastic syndromes. Leukemia 2012; 27:610-8. [PMID: 22936014 DOI: 10.1038/leu.2012.253] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene expression profiling signatures may be used to classify the subtypes of Myelodysplastic syndrome (MDS) patients. However, there are few reports on the global methylation status in MDS. The integration of genome-wide epigenetic regulatory marks with gene expression levels would provide additional information regarding the biological differences between MDS and healthy controls. Gene expression and methylation status were measured using high-density microarrays. A total of 552 differentially methylated CpG loci were identified as being present in low-risk MDS; hypermethylated genes were more frequent than hypomethylated genes. In addition, mRNA expression profiling identified 1005 genes that significantly differed between low-risk MDS and the control group. Integrative analysis of the epigenetic and expression profiles revealed that 66.7% of the hypermethylated genes were underexpressed in low-risk MDS cases. Gene network analysis revealed molecular mechanisms associated with the low-risk MDS group, including altered apoptosis pathways. The two key apoptotic genes BCL2 and ETS1 were identified as silenced genes. In addition, the immune response and micro RNA biogenesis were affected by the hypermethylation and underexpression of IL27RA and DICER1. Our integrative analysis revealed that aberrant epigenetic regulation is a hallmark of low-risk MDS patients and could have a central role in these diseases.
Collapse
Affiliation(s)
- M del Rey
- Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC, Salamanca, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
430
|
Kim TS, Lee DH, Kim SK, Shin SY, Seo EJ, Lim DS. Mammalian sterile 20-like kinase 1 suppresses lymphoma development by promoting faithful chromosome segregation. Cancer Res 2012; 72:5386-95. [PMID: 22926556 DOI: 10.1158/0008-5472.can-11-3956] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The mammalian Hippo signaling pathway has been implicated in oncogenesis in the context of solid tumors such as hepatocellular carcinoma. Mammalian sterile 20-like kinase 1 (MST1), the core component of the Hippo signaling pathway, is highly expressed in hematopoietic cells. However, its possible impact on tumorigenesis in this setting is unknown. In this study, we provide evidence that Mst1 loss in the mouse enhances chemically and genetically induced lymphoma development by inducing chromosomal instability. Mst1 deficiency increased susceptibility to T-cell acute lymphoblastic leukemia induced by mutagen exposure. Notably, before transformation Mst1(-/-) normal thymocytes showed no changes in proliferation or apoptosis in vitro and in vivo, but they displayed elevated levels of abnormal mitotic chromosomes and aneuploidy, conditions known to promote tumorigenesis. Mst1(-/-) mice also showed accelerated formation of spontaneous lymphomas in a p53-deficient background, accompanied by severe aneuploidy. In clinical specimens of lymphoma and leukemia, we documented frequent downregulation of MST1 expression, consistent with our findings. Taken together, our findings reveal a tumor suppressive function of Mst1 based on its ability to prevent chromosomal instability in lymphocytes.
Collapse
Affiliation(s)
- Tae-Shin Kim
- Authors' Affiliations: Department of Biological Sciences, National Creative Research Initiatives Center, Graduate School of Nanoscience and Technology (WCU), Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | | | | | | | | | | |
Collapse
|
431
|
Khan I, Altman JK, Licht JD. New strategies in acute myeloid leukemia: redefining prognostic markers to guide therapy. Clin Cancer Res 2012; 18:5163-71. [PMID: 22893630 DOI: 10.1158/1078-0432.ccr-12-0313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Although standard therapy for AML has been relatively constant over the past 2 decades, this may be changing with enhanced technologies allowing for the classification of acute myeloid leukemia (AML) into molecularly distinct subsets. Some specific subsets of AML have an excellent prognosis in response to standard therapy, whereas the poor prognosis of AML associated with specific sets of mutations or chromosomal anomalies requires the development of new therapies. Elucidation of the molecular pathogenesis of AML has led to the development of therapies that affect signaling, apoptosis, protein and intermediate metabolism, the surface of the leukemia cell, leukemia cell/stromal interaction, and epigenetic regulation of gene expression.
Collapse
Affiliation(s)
- Irum Khan
- Division of Hematology/Oncology, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
432
|
Subnetwork-based analysis of chronic lymphocytic leukemia identifies pathways that associate with disease progression. Blood 2012; 120:2639-49. [PMID: 22837534 DOI: 10.1182/blood-2012-03-416461] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The clinical course of patients with chronic lymphocytic leukemia (CLL) is heterogeneous. Several prognostic factors have been identified that can stratify patients into groups that differ in their relative tendency for disease progression and/or survival. Here, we pursued a subnetwork-based analysis of gene expression profiles to discriminate between groups of patients with disparate risks for CLL progression. From an initial cohort of 130 patients, we identified 38 prognostic subnetworks that could predict the relative risk for disease progression requiring therapy from the time of sample collection, more accurately than established markers. The prognostic power of these subnetworks then was validated on 2 other cohorts of patients. We noted reduced divergence in gene expression between leukemia cells of CLL patients classified at diagnosis with aggressive versus indolent disease over time. The predictive subnetworks vary in levels of expression over time but exhibit increased similarity at later time points before therapy, suggesting that degenerate pathways apparently converge into common pathways that are associated with disease progression. As such, these results have implications for understanding cancer evolution and for the development of novel treatment strategies for patients with CLL.
Collapse
|
433
|
Iacobucci I, Iraci N, Messina M, Lonetti A, Chiaretti S, Valli E, Ferrari A, Papayannidis C, Paoloni F, Vitale A, Storlazzi CT, Ottaviani E, Guadagnuolo V, Durante S, Vignetti M, Soverini S, Pane F, Foà R, Baccarani M, Müschen M, Perini G, Martinelli G. IKAROS deletions dictate a unique gene expression signature in patients with adult B-cell acute lymphoblastic leukemia. PLoS One 2012; 7:e40934. [PMID: 22848414 PMCID: PMC3405023 DOI: 10.1371/journal.pone.0040934] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 06/15/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Deletions of IKAROS (IKZF1) frequently occur in B-cell precursor acute lymphoblastic leukemia (B-ALL) but the mechanisms by which they influence pathogenesis are unclear. To address this issue, a cohort of 144 adult B-ALL patients (106 BCR-ABL1-positive and 38 B-ALL negative for known molecular rearrangements) was screened for IKZF1 deletions by single nucleotide polymorphism (SNP) arrays; a sub-cohort of these patients (44%) was then analyzed for gene expression profiling. PRINCIPAL FINDINGS Total or partial deletions of IKZF1 were more frequent in BCR-ABL1-positive than in BCR-ABL1-negative B-ALL cases (75% vs 58%, respectively, p = 0.04). Comparison of the gene expression signatures of patients carrying IKZF1 deletion vs those without showed a unique signature featured by down-regulation of B-cell lineage and DNA repair genes and up-regulation of genes involved in cell cycle, JAK-STAT signalling and stem cell self-renewal. Through chromatin immunoprecipitation and luciferase reporter assays we corroborated these findings both in vivo and in vitro, showing that Ikaros deleted isoforms lacked the ability to directly regulate a large group of the genes in the signature, such as IGLL1, BLK, EBF1, MSH2, BUB3, ETV6, YES1, CDKN1A (p21), CDKN2C (p18) and MCL1. CONCLUSIONS Here we identified and validated for the first time molecular pathways specifically controlled by IKZF1, shedding light into IKZF1 role in B-ALL pathogenesis.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Nunzio Iraci
- Department of Biology, University of Bologna, Bologna, Italy
| | - Monica Messina
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, “Sapienza” University of Rome, Rome, Italy
| | - Annalisa Lonetti
- Cellular Signalling Laboratory, Department of Human Anatomy, University of Bologna, Bologna, Italy
| | - Sabina Chiaretti
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, “Sapienza” University of Rome, Rome, Italy
| | - Emanuele Valli
- Department of Biology, University of Bologna, Bologna, Italy
| | - Anna Ferrari
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Cristina Papayannidis
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Francesca Paoloni
- Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto (GIMEMA) Data Center, GIMEMA Foundation, Rome, Italy
| | - Antonella Vitale
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, “Sapienza” University of Rome, Rome, Italy
| | | | - Emanuela Ottaviani
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Viviana Guadagnuolo
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Sandra Durante
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Marco Vignetti
- Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto (GIMEMA) Data Center, GIMEMA Foundation, Rome, Italy
| | - Simona Soverini
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Fabrizio Pane
- CEINGE Biotecnologie Avanzate and Department of Biochemistry and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Robin Foà
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, “Sapienza” University of Rome, Rome, Italy
| | - Michele Baccarani
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Markus Müschen
- Leukemia Research Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California, United States of America
| | - Giovanni Perini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- Department of Hematology and Oncological Sciences “L. and A. Seràgnoli”, S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
- * E-mail:
| |
Collapse
|
434
|
Murati A, Brecqueville M, Devillier R, Mozziconacci MJ, Gelsi-Boyer V, Birnbaum D. Myeloid malignancies: mutations, models and management. BMC Cancer 2012; 12:304. [PMID: 22823977 PMCID: PMC3418560 DOI: 10.1186/1471-2407-12-304] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 06/30/2012] [Indexed: 12/05/2022] Open
Abstract
Myeloid malignant diseases comprise chronic (including myelodysplastic syndromes, myeloproliferative neoplasms and chronic myelomonocytic leukemia) and acute (acute myeloid leukemia) stages. They are clonal diseases arising in hematopoietic stem or progenitor cells. Mutations responsible for these diseases occur in several genes whose encoded proteins belong principally to five classes: signaling pathways proteins (e.g. CBL, FLT3, JAK2, RAS), transcription factors (e.g. CEBPA, ETV6, RUNX1), epigenetic regulators (e.g. ASXL1, DNMT3A, EZH2, IDH1, IDH2, SUZ12, TET2, UTX), tumor suppressors (e.g. TP53), and components of the spliceosome (e.g. SF3B1, SRSF2). Large-scale sequencing efforts will soon lead to the establishment of a comprehensive repertoire of these mutations, allowing for a better definition and classification of myeloid malignancies, the identification of new prognostic markers and therapeutic targets, and the development of novel therapies. Given the importance of epigenetic deregulation in myeloid diseases, the use of drugs targeting epigenetic regulators appears as a most promising therapeutic approach.
Collapse
Affiliation(s)
- Anne Murati
- Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Oncologie Moléculaire; UMR1068 Inserm, Institut Paoli-Calmettes, 27 Bd, Leï Roure, BP 30059, Marseille, 13273, France
| | | | | | | | | | | |
Collapse
|
435
|
Hannon MM, Lohan F, Erbilgin Y, Sayitoglu M, O'Hagan K, Mills K, Ozbek U, Keeshan K. Elevated TRIB2 with NOTCH1 activation in paediatric/adult T-ALL. Br J Haematol 2012; 158:626-34. [PMID: 22775572 DOI: 10.1111/j.1365-2141.2012.09222.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 05/17/2012] [Indexed: 01/22/2023]
Abstract
TRIB2 is a potent oncogene, elevated in a subset of human acute myeloid leukaemias (AML) with a mixed myeloid/lymphoid phenotype and NOTCH1 mutations. Although rare in AML, activating NOTCH1 mutations occur in 50% of all T cell acute lymphoblastic leukaemias (T-ALL). TRIB2 is a NOTCH1 target gene that functions in the degradation of key proteins and modulation of MAPK signalling pathways, implicated in haematopoietic cell survival and proliferation. This study showed that TRIB2 expression level is highest in the lymphoid compartment of normal haematopoietic cells, specifically in T cells. Analysis of TRIB2 expression across 16 different subtypes of human leukaemia demonstrated that TRIB2 expression was higher in ALL phenotypes versus all other phenotypes including AML, chronic lymphocytic leukaemia (CLL), myelodysplastic syndrome (MDS) and chronic myeloid leukaemia (CML). A T cell profile was distinguished by high TRIB2 expression in normal and malignant haematopoiesis. High TRIB2 expression was seen in T-ALL with normal karyotype and correlated with NOTCH signalling pathways. High TRIB2 expression correlated with NOTCH1/FBXW7 mutations in a paediatric T-ALL cohort, strongly linking NOTCH1 activation and high TRIB2 expression in paediatric T-ALL. The relationship between TRIB2 and T cell signalling pathways uniquely identifies leukaemia subtypes and will be useful in the advancement of our understanding of T cell and ALL biology.
Collapse
Affiliation(s)
- Maura M Hannon
- Department of Biochemsitry, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
436
|
Wernicke CM, Richter GHS, Beinvogl BC, Plehm S, Schlitter AM, Bandapalli OR, Prazeres da Costa O, Hattenhorst UE, Volkmer I, Staege MS, Esposito I, Burdach S, Grunewald TGP. MondoA is highly overexpressed in acute lymphoblastic leukemia cells and modulates their metabolism, differentiation and survival. Leuk Res 2012; 36:1185-92. [PMID: 22748921 DOI: 10.1016/j.leukres.2012.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 01/28/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. To identify novel candidates for targeted therapy, we performed a comprehensive transcriptome analysis identifying MondoA (MLXIP) - a transcription factor regulating glycolysis - to be overexpressed in ALL compared to normal tissues. Using microarray-profiling, gene-set enrichment analysis, RNA interference and functional assays we show that MondoA overexpression increases glucose catabolism and maintains a more immature phenotype, which is associated with enhanced survival and clonogenicity of leukemia cells. These data point to an important contribution of MondoA to leukemia aggressiveness and make MondoA a potential candidate for targeted treatment of ALL.
Collapse
Affiliation(s)
- Caroline M Wernicke
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Laboratory of Functional Genomics and Transplantation Biology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
437
|
HemaExplorer: a Web server for easy and fast visualization of gene expression in normal and malignant hematopoiesis. Blood 2012; 119:6394-5. [PMID: 22745298 DOI: 10.1182/blood-2012-05-427310] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
438
|
Leukemia gene atlas--a public platform for integrative exploration of genome-wide molecular data. PLoS One 2012; 7:e39148. [PMID: 22720055 PMCID: PMC3375295 DOI: 10.1371/journal.pone.0039148] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/16/2012] [Indexed: 11/19/2022] Open
Abstract
Leukemias are exceptionally well studied at the molecular level and a wealth of high-throughput data has been published. But further utilization of these data by researchers is severely hampered by the lack of accessible integrative tools for viewing and analysis. We developed the Leukemia Gene Atlas (LGA) as a public platform designed to support research and analysis of diverse genomic data published in the field of leukemia. With respect to leukemia research, the LGA is a unique resource with comprehensive search and browse functions. It provides extensive analysis and visualization tools for various types of molecular data. Currently, its database contains data from more than 5,800 leukemia and hematopoiesis samples generated by microarray gene expression, DNA methylation, SNP and next generation sequencing analyses. The LGA allows easy retrieval of large published data sets and thus helps to avoid redundant investigations. It is accessible at www.leukemia-gene-atlas.org.
Collapse
|
439
|
Laffin B, Petrash JM. Expression of the Aldo-Ketoreductases AKR1B1 and AKR1B10 in Human Cancers. Front Pharmacol 2012; 3:104. [PMID: 22685431 PMCID: PMC3368246 DOI: 10.3389/fphar.2012.00104] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/13/2012] [Indexed: 01/27/2023] Open
Abstract
The American Cancer Society estimates that there will be more than 1.5 million new cases of cancer in 2011, underscoring the need for identification of new therapeutic targets and development of novel cancer therapies. Previous studies have implicated the human aldo-ketoreductases AKR1B1 and AKR1B10 in cancer, and therefore we examined AKR1B1 and AKR1B10 expression across all major human cancer types using the Oncomine cancer gene expression database (Compendia Biosciences, www.oncomine.com). Using this database, we found that expression of AKR1B1 and AKR1B10 varies greatly by cancer type and tissue of origin, including agreement with previous reports that AKR1B10 is significantly over-expressed in cancers of the lungs and liver. AKR1B1 is more broadly over-expressed in human cancers than AKR1B10, albeit at a generally lower magnitude. AKR1B1 over-expression was found to be associated with shortened patient survival in acute myelogenous leukemias and multiple myelomas. High AKR1B10 expression tends to predict less aggressive clinical course generally, notably within lung cancers, where it tends to be highly over-expressed compared to normal tissue. These findings suggest that AKR1B1 inhibitors in particular hold great potential as novel cancer therapeutics.
Collapse
Affiliation(s)
- Brian Laffin
- Department of Ophthalmology, The School of Medicine, University of Colorado Aurora, CO, USA
| | | |
Collapse
|
440
|
Abstract
Abstract
Osteopontin (OPN) is a glycoprotein that is secreted by osteoblasts and hematopoietic cells. OPN suppresses the proliferation of hematopoietic stem cells in vitro and may regulate the hematopoietic stem cell pool. Increased serum OPN concentrations occur in chronic myeloid leukemia, multiple myeloma, and acute myeloid leukemia (AML). In the present study, we analyzed the prognostic impact of OPN in AML by investigating the expression and relevance of OPN in newly diagnosed AML patients from 2 large study groups (the German AML Cooperative Group and the Dutch-Belgian Hematology Oncology Cooperative group). IHC (n = 84), ELISAs of blood/BM sera (n = 41), and microarray data for mRNA levels (n = 261) were performed. Expression of OPN protein was increased in AML patients both in BM blasts (IHC) and in BM serum (ELISA) compared with healthy controls. Patients expressing high levels of OPN within the BM (IHC) experienced shortened overall survival (OS; P = .025). Multivariate analysis identified karyotype, blast clearance (day 16), and the level of OPN expression as independent prognostic factors for OS. This prompted us to analyze microarray data from 261 patients from a third cohort. The analysis confirmed OPN as a prognostic marker. In summary, high OPN mRNA expression indicated decreased event-free survival (P = .0002) and OS (P = .001). The prognostic role of OPN was most prominent in intermediate-risk AML. These data provide evidence that OPN expression is an independent prognostic factor in AML.
Collapse
|
441
|
Federzoni EA, Valk PJM, Torbett BE, Haferlach T, Löwenberg B, Fey MF, Tschan MP. PU.1 is linking the glycolytic enzyme HK3 in neutrophil differentiation and survival of APL cells. Blood 2012; 119:4963-70. [PMID: 22498738 PMCID: PMC3367898 DOI: 10.1182/blood-2011-09-378117] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 02/08/2012] [Indexed: 11/20/2022] Open
Abstract
The transcription factor PU.1 is a master regulator of myeloid differentiation and function. On the other hand, only scarce information is available on PU.1-regulated genes involved in cell survival. We now identified the glycolytic enzyme hexokinase 3 (HK3), a gene with cytoprotective functions, as transcriptional target of PU.1. Interestingly, HK3 expression is highly associated with the myeloid lineage and was significantly decreased in acute myeloid leukemia patients compared with normal granulocytes. Moreover, HK3 expression was significantly lower in acute promyelocytic leukemia (APL) compared with non-APL patient samples. In line with the observations in primary APL patient samples, we observed significantly higher HK3 expression during neutrophil differentiation of APL cell lines. Moreover, knocking down PU.1 impaired HK3 induction during neutrophil differentiation. In vivo binding of PU.1 and PML-RARA to the HK3 promoter was found, and PML-RARA attenuated PU.1 activation of the HK3 promoter. Next, inhibiting HK3 in APL cell lines resulted in significantly reduced neutrophil differentiation and viability compared with control cells. Our findings strongly suggest that HK3 is: (1) directly activated by PU.1, (2) repressed by PML-RARA, and (3) functionally involved in neutrophil differentiation and cell viability of APL cells.
Collapse
MESH Headings
- Anthracyclines/pharmacology
- Anthracyclines/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Survival/drug effects
- Cell Survival/genetics
- Cells, Cultured
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/physiology
- Glycolysis/genetics
- Hexokinase/genetics
- Hexokinase/metabolism
- Hexokinase/physiology
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Neutrophils/drug effects
- Neutrophils/metabolism
- Neutrophils/pathology
- Neutrophils/physiology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/physiology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Trans-Activators/physiology
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
Collapse
Affiliation(s)
- Elena A Federzoni
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
442
|
Rager JE, Fry RC. The aryl hydrocarbon receptor pathway: a key component of the microRNA-mediated AML signalisome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2012; 9:1939-53. [PMID: 22754483 PMCID: PMC3386597 DOI: 10.3390/ijerph9051939] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/27/2012] [Accepted: 05/08/2012] [Indexed: 11/16/2022]
Abstract
Recent research has spotlighted the role of microRNAs (miRNAs) as critical epigenetic regulators of hematopoietic stem cell differentiation and leukemia development. Despite the recent advances in knowledge surrounding epigenetics and leukemia, the mechanisms underlying miRNAs' influence on leukemia development have yet to be clearly elucidated. Our aim was to identify high ranking biological pathways altered at the gene expression level and under epigenetic control. Specifically, we set out to test the hypothesis that miRNAs dysregulated in acute myeloid leukemia (AML) converge on a common pathway that can influence signaling related to hematopoiesis and leukemia development. We identified genes altered in AML patients that are under common regulation of seven key miRNAs. By mapping these genes to a global interaction network, we identified the "AML Signalisome". The AML Signalisome comprises 53 AML-associated molecules, and is enriched for proteins that play a role in the aryl hydrocarbon receptor (AhR) pathway, a major regulator of hematopoiesis. Furthermore, we show biological enrichment for hematopoiesis-related proteins within the AML Signalisome. These findings provide important insight into miRNA-regulated pathways in leukemia, and may help to prioritize targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Julia E. Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina, 135 Dauer Drive, CB 7431, UNC, Chapel Hill, NC 27599, USA;
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina, 135 Dauer Drive, CB 7431, UNC, Chapel Hill, NC 27599, USA;
- Curriculum in Toxicology, The University of North Carolina, 135 Dauer Drive, CB 7431, UNC, Chapel Hill, NC 27599, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-919-843-6864; Fax: +1-919-843-9047
| |
Collapse
|
443
|
Beisser D, Brunkhorst S, Dandekar T, Klau GW, Dittrich MT, Müller T. Robustness and accuracy of functional modules in integrated network analysis. ACTA ACUST UNITED AC 2012; 28:1887-94. [PMID: 22581175 DOI: 10.1093/bioinformatics/bts265] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
MOTIVATION High-throughput molecular data provide a wealth of information that can be integrated into network analysis. Several approaches exist that identify functional modules in the context of integrated biological networks. The objective of this study is 2-fold: first, to assess the accuracy and variability of identified modules and second, to develop an algorithm for deriving highly robust and accurate solutions. RESULTS In a comparative simulation study accuracy and robustness of the proposed and established methodologies are validated, considering various sources of variation in the data. To assess this variation, we propose a jackknife resampling procedure resulting in an ensemble of optimal modules. A consensus approach summarizes the ensemble into one final module containing maximally robust nodes and edges. The resulting consensus module identifies and visualizes robust and variable regions by assigning support values to nodes and edges. Finally, the proposed approach is exemplified on two large gene expression studies: diffuse large B-cell lymphoma and acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Daniela Beisser
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
444
|
Nordlund J, Milani L, Lundmark A, Lönnerholm G, Syvänen AC. DNA methylation analysis of bone marrow cells at diagnosis of acute lymphoblastic leukemia and at remission. PLoS One 2012; 7:e34513. [PMID: 22493696 PMCID: PMC3321015 DOI: 10.1371/journal.pone.0034513] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/06/2012] [Indexed: 01/17/2023] Open
Abstract
To detect genes with CpG sites that display methylation patterns that are characteristic of acute lymphoblastic leukemia (ALL) cells, we compared the methylation patterns of cells taken at diagnosis from 20 patients with pediatric ALL to the methylation patterns in mononuclear cells from bone marrow of the same patients during remission and in non-leukemic control cells from bone marrow or blood. Using a custom-designed assay, we measured the methylation levels of 1,320 CpG sites in regulatory regions of 413 genes that were analyzed because they display allele-specific gene expression (ASE) in ALL cells. The rationale for our selection of CpG sites was that ASE could be the result of allele-specific methylation in the promoter regions of the genes. We found that the ALL cells had methylation profiles that allowed distinction between ALL cells and control cells. Using stringent criteria for calling differential methylation, we identified 28 CpG sites in 24 genes with recurrent differences in their methylation levels between ALL cells and control cells. Twenty of the differentially methylated genes were hypermethylated in the ALL cells, and as many as nine of them (AMICA1, CPNE7, CR1, DBC1, EYA4, LGALS8, RYR3, UQCRFS1, WDR35) have functions in cell signaling and/or apoptosis. The methylation levels of a subset of the genes were consistent with an inverse relationship with the mRNA expression levels in a large number of ALL cells from published data sets, supporting a potential biological effect of the methylation signatures and their application for diagnostic purposes.
Collapse
Affiliation(s)
- Jessica Nordlund
- Department of Medical Sciences, Molecular Medicine, Uppsala University, Uppsala, Sweden
| | - Lili Milani
- Department of Medical Sciences, Molecular Medicine, Uppsala University, Uppsala, Sweden
| | - Anders Lundmark
- Department of Medical Sciences, Molecular Medicine, Uppsala University, Uppsala, Sweden
| | - Gudmar Lönnerholm
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Ann-Christine Syvänen
- Department of Medical Sciences, Molecular Medicine, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
445
|
McGregor S, McNeer J, Gurbuxani S. Beyond the 2008 World Health Organization classification: the role of the hematopathology laboratory in the diagnosis and management of acute lymphoblastic leukemia. Semin Diagn Pathol 2012; 29:2-11. [PMID: 22372201 DOI: 10.1053/j.semdp.2011.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The diagnosis of acute lymphoblastic leukemia (ALL) is made by evaluating morphology and immunophenotype. However, appropriate risk stratification and decisions regarding the intensity of therapy are influenced by additional clinical and laboratory testing that reflect the biology of the disease. Recent years have seen tremendous progress in uncovering genetic lesions that influence the biology of ALL. In recognition of these advances, the 2008 WHO classification incorporated the category of B-lymphoblastic leukemia/lymphoma with recurrent genetic abnormalities into the classification of precursor lymphoid neoplasms. Based on the knowledge available at the time, genetic lesions associated with distinct clinical features, immunophenotype, prognosis, or other unique biological characteristics were included in this category. Not surprisingly, significant novel genetic lesions that profoundly affect the biology of ALL have since been identified and will have a major impact on risk stratification and may ultimately be incorporated into future classification schemes. After establishing an initial diagnosis and treatment regimen, hematopathologists must also evaluate for minimal residual disease (MRD) to determine the need for additional intervention because MRD remains the most useful clinical indicator of disease progression and response to treatment. Doing so requires familiarity with not only morphology, but also flow cytometry and molecular genetics. Although not all of these applications are handled directly by the hematopathologist, it is our strong belief that meaningful involvement in patient care dictates that hematopathologists appreciate all aspects of ALL diagnosis and disease monitoring. This review covers the salient aspects of recent advances in the biology of ALL and evaluation of MRD, placing emphasis on how this information may ultimately be used to improve risk stratification and, as a result, patient outcomes.
Collapse
Affiliation(s)
- Stephanie McGregor
- Department of Pathology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
446
|
Bou Samra E, Moreaux J, Vacheret F, Mills K, Rufflé F, Chiesa J, Piquemal D, Boureux A, Lavabre-Bertrand T, Jourdan E, Commes T. New prognostic markers, determined using gene expression analyses, reveal two distinct subtypes of chronic myelomonocytic leukaemia patients. Br J Haematol 2012; 157:347-56. [PMID: 22390678 DOI: 10.1111/j.1365-2141.2012.09069.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/16/2012] [Indexed: 01/25/2023]
Abstract
Chronic myelomonocytic leukaemia (CMML) is a heterogeneous haematopoietic disorder characterized by myeloproliferative or myelodysplastic features. At present, the pathogenesis of this malignancy is not completely understood. In this study, we sought to analyse gene expression profiles of CMML in order to characterize new molecular outcome predictors. A learning set of 32 untreated CMML patients at diagnosis was available for TaqMan low-density array gene expression analysis. From 93 selected genes related to cancer and cell cycle, we built a five-gene prognostic index after multiplicity correction. Using this index, we characterized two categories of patients with distinct overall survival (94% vs. 19% for good and poor overall survival, respectively; P = 0·007) and we successfully validated its strength on an independent cohort of 21 CMML patients with Affymetrix gene expression data. We found no specific patterns of association with traditional prognostic stratification parameters in the learning cohort. However, the poor survival group strongly correlated with high-risk treated patients and transformation to acute myeloid leukaemia. We report here a new multigene prognostic index for CMML, independent of the gene expression measurement method, which could be used as a powerful tool to predict clinical outcome and help physicians to evaluate criteria for treatments.
Collapse
Affiliation(s)
- Elias Bou Samra
- Groupe d'études des transcriptomes, Université Montpellier 2, Centre de Recherche en Biochimie Macromoléculaire, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
447
|
Ren K, Lu J, Porollo A, Du C. Tumor-suppressing function of caspase-2 requires catalytic site Cys-320 and site Ser-139 in mice. J Biol Chem 2012; 287:14792-802. [PMID: 22396545 DOI: 10.1074/jbc.m112.347625] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The multifunctional caspase-2 protein is involved in apoptosis, NF-κB regulation, and tumor suppression in mice. However, the mechanisms of caspase-2 responsible for tumor suppression remain unclear. Here we identified two sites of caspase-2, the catalytic Cys-320 site and the Ser-139 site, to be important for suppression of cellular transformation and tumorigenesis. Using SV40- and K-Ras-transformed caspase-2 KO mouse embryonic fibroblast cells reconstituted with expression of wild-type, catalytic dead (C320A), or Ser-139 (S139A) mutant caspase-2, we demonstrated that similar to caspase-2 deficiency, when Cys-320 and Ser-139 were mutated, caspase-2 lost its ability to inhibit cellular transformation and tumorigenesis. These mutant cells exhibited enhanced cell proliferation, elevated clonogenic activity, accelerated anchorage-independent growth, and transformation and were highly tumorigenic, rapidly producing large tumors in athymic nude mice. Investigation into the underlying mechanism showed that these two residues are needed for caspase-2 to suppress NF-κB activity, promote apoptosis, and sustain the G(2)/M checkpoint following DNA damage induction. In addition, tumors in nude mice derived from the two mutant cell lines had higher constitutive NF-κB activity and elevated expression of NF-κB targets of antiapoptotic proteins Bcl-xL, XIAP, and cIAP2. A reduction in caspase-2 mRNA was associated with multiple types of cancers in patients. Together, these observations suggest the combined functions of caspase-2 in suppressing NF-κB activation, promoting apoptosis, and sustaining G(2)/M checkpoint contribute to caspase-2 tumor-suppressing function and that caspase-2 may also impact tumor suppression in humans. These findings provide insight into tumor suppression at the cross-roads of apoptosis, cell cycle checkpoint, and NF-κB pathways.
Collapse
Affiliation(s)
- Keqin Ren
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | | | | | | |
Collapse
|
448
|
Bacalini MG, Tavolaro S, Peragine N, Marinelli M, Santangelo S, Del Giudice I, Mauro FR, Di Maio V, Ricciardi MR, Caiafa P, Chiaretti S, Foà R, Guarini A, Reale A. A subset of chronic lymphocytic leukemia patients display reduced levels of PARP1 expression coupled with a defective irradiation-induced apoptosis. Exp Hematol 2012; 40:197-206.e1. [DOI: 10.1016/j.exphem.2011.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 10/25/2011] [Accepted: 11/14/2011] [Indexed: 02/03/2023]
|
449
|
Oh S, Janknecht R. Histone demethylase JMJD5 is essential for embryonic development. Biochem Biophys Res Commun 2012; 420:61-5. [DOI: 10.1016/j.bbrc.2012.02.115] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 02/21/2012] [Indexed: 01/08/2023]
|
450
|
Mato A, Feldman T, Richter J, Siegel DS, Goy A. Liquid tumors in the elderly. Clin Geriatr Med 2012; 28:115-52. [PMID: 22326039 DOI: 10.1016/j.cger.2011.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Anthony Mato
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA.
| | | | | | | | | |
Collapse
|