scholarly journals The Condensin II Complex Subunit NCAPH2 Is Essential for Erythropoiesis and Regulates Erythroid Gene Expression

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 286-286
Author(s):  
Zachary C. Murphy ◽  
Maeve Wells ◽  
Kristin Murphy ◽  
Michael Getman ◽  
Laurie A. Steiner

Abstract Erythropoiesis requires dramatic changes in gene expression in a cell that is rapidly proliferating and undergoing progressive nuclear condensation in anticipation of enucleation. Disruption of this process is associated with myelodysplastic syndromes and congenital anemias. Our lab has demonstrated that Setd8, the sole histone methyltransferase that can generate H4K20me1, plays an essential role in this process (Malik 2019). H4K20me1 accumulates during terminal erythroid maturation (Murphy Blood 2021) and can regulate chromatin structure and gene expression through interaction with multiple partners, including the Condensin II Complex. The Condensin II complex is a ring-like structure composed of two conserved SMC components (SMC2 and SMC4), two HEAT subunits (NCAPG2 and NCAPD3), and a kleisin subunit NCAPH2. The Condensin II complex plays an important role in chromatin condensation during mitosis, and establishing higher-order chromatin interactions in interphase cells, with some studies suggesting it also regulates gene expression (Yuen Science Adv 2017; Iwasaki Nature Comm 2019). Similar to Setd8, many subunits of the Condensin II complex are highly expressed in erythroid cells compared to most other cell types (biogps.org). We hypothesized that the Condensin II complex, in conjunction with Setd8 and H4K20me1, is important for establishing appropriate patterns of chromatin architecture and gene expression in maturing erythroblasts. To address this hypothesis, we deleted the NCAPH2 subunit in erythroid cells by crossing mice with floxed alleles of NCAPH2 with mice expressing cre-recombinase under the direction of the Erythropoietin receptor promotor (EpoRCre). Homozygous disruption of NCAPH2 was embryonic lethal by E13.5. NCAPH2 mutant embryos were similar in appearance to littermate controls until E12.5 when they developed notable pallor and a dramatic decline in the number of benzidine positive cells. Cell cycle analyses demonstrated that an accumulation of cells in G2/M preceded the dramatic decline in erythroblast numbers at E12.5. In contrast to cells from littermate controls, the NCAPH2 mutant cells were very heterogenous in cell and nuclear size and morphology. Surpisingly, most NCAPH2 mutant cells appeared to be hemoglobinized, suggesting sufficient iron accumulation and heme synthesis. In vitro cultures derived from primitive erythroid progenitors replicated in vivo findings, including normal early erythropoeisis, with significant abnormalities during mid- to late- maturation. Western blot in cycloheximide treated primitive erythroid cultures revealed that NCAPH2 has a long half-life, which likely contributes to the relative normalicy of early primitive erythoproesis. NCAPH2 mutant embryos also had a dramatic failure of definitive erythropoiesis, as evidenced by loss of mature erythroblasts in the fetal liver at E13.5. To gain insights into the mechanisms underlying these findings, we performed RNA-seq of NCAPH2 mutant, NCAPH2 het, and NCAPH2 WT erythroblasts from E11.5 embryos. Comparing NCAPH2 mutant and NCAPH2 WT erythroblasts there were 1121 down regulated genes and 743 upregulated genes (adj p-value <0.05). As expected, the downregulated genes were enriched for pathways related to cell cycle, such as Mitotic Spindle Organization (adj pvalue 5e-42). The upregulated genes were enriched for a variety of pathways including p53 transcriptional network (adj pvalue 4e-10), neutrophil mediated immunity (2e-9), DNA-binding transcription factor (adj pvalue 7e-5), and regulation of erythrocyte differentiation (adj pvalue 5e-4). Intriguingly, 91/340 genes differentially expressed in Setd8 mutant erythroblats were also differentially expressed in NCAPH2 mutant cells, including genes typically repressed early in erythroid commitment, such as GATA2 and SPI1. Cut&Tag in CD34+ derived erythroblasts demonstrated occupancy of H4K20me1 at these loci. Mass spectrometry of proteins isolated via mono-mehtylated H4K20 peptides in erythroid extracts identified Condensin II components, supporting a model where the Condensin II complex directly interacts with H4K20me1. Together, these results demonstrate that the Condensin II complex is essential for erythropoiesis, and may work in conjunction with Setd8 and H4K20me1 to establish appropriate patterns of gene expression in maturing erythroblasts. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 499-499
Author(s):  
Tyler A Couch ◽  
Zachary C. Murphy ◽  
Jacquelyn Lillis ◽  
Michael Getman ◽  
Paul D. Kingsley ◽  
...  

Abstract Setd8 is the sole histone methyltransferase capable of mono-methylating histone H4, lysine 20. Setd8 is expressed at basal levels in most cell types and is important for many basic cellular functions, including cell cycle progression, transcriptional regulation, and mitotic chromatin condensation. Setd8 is expressed ~10-fold higher in erythroblasts than any other cell type and during erythroid maturation of human CD34+ HSPC, Setd8 protein levels increase in parallel with Gata1 levels, suggesting that Setd8 may have an erythroid-specific function(s). Consistent with this hypothesis, erythroid-specific deletion of Setd8 was embryonic lethal, resulting in profound anemia. Setd8-null erythroblasts had cell cycle abnormalities, failure of transcriptional repression, and defective terminal erythroid maturation. (Malik et al., Cell Reports, 2017). These studies provided important insights into the function of Setd8 in erythroid cells, but were not able to clearly delineate the "housekeeping" functions of Setd8 from its specific functions in erythropoiesis. To identify the erythroid-specific functions of Setd8, we sought to identify and disrupt the enhancer that drives high level Setd8 expression in erythroid cells. Using publically available ChIP-seq data sets, we identified a putative enhancer located in intron 1 of the SETD8 gene that was occupied by Gata1, Tal1, and H3K4me1 in human erythroblasts derived from culture of CD43+ HSPCs. This putative enhancer was able to drive luciferase expression in a reporter gene assay, and deletion of the Gata1:Tal1 site at the center of this region was sufficient to abrogate reporter gene activity. Based on these data, we hypothesized that this was the enhancer that drives high level expression of Setd8 in erythroid cells. To test this hypothesis, we used CRISPR/Cas9 genome editing to delete this region in HUDEP-2 cells. Briefly, Cas9 and guide RNA ribonucleoprotein complexes targeting the enhancer were delivered into the cells using electroporation (Gundry et al., Cell Reports, 2015). PCR and sequencing were used to confirm genome editing in monoclonal cell lines. Homozygous deletion of the enhancer (Δ/Δ) reduced SETD8 expression to 27.8% of WT (+/+) controls by RT-qPCR (n=3 for each genotype; p=0.0018). Decreased Setd8 protein levels and H4K20 mono-methylation was confirmed by Western blot. Further supporting an important function of Setd8 in erythropoiesis, deletion of the enhancer and exon 7 in CD34+ HSPCs resulted in a decreased efficiency of erythroid colony formation to 49.6% of control (n=5, p=0.0359). To gain insights into Setd8 gene regulation in erythroid cells, we performed RNA-seq, comparing the Δ/Δ and +/+ enhancer lines. In total, there were 603 genes differentially expressed (p<0.05; fold change >1.5), including SETD8, FAS, and CDKN1A (p21Cip1). Pathway analyses identified numerous genes associated with apoptosis and cell death to be up-regulated. Intriguingly, multiple genes in important for stress erythropoiesis were differentially expressed in the Setd8 Δ/Δ and +/+ enhancer lines and were also differentially expressed in Setd8-null murine erythroblasts (Malik et al., Cell Reports, 2017). Most notably, both the Δ/Δ enhancer lines and the Setd8-null erythroblasts had significantly higher levels of Fas death receptor transcript than control cells. Down-regulation of Fas is essential for stress erythropoiesis (Liu et al., Blood, 2006). We therefore hypothesized that Setd8 is important for the stress erythropoiesis response. To test this hypothesis, we subjected EpoR-Cre+/-;Setd8fl/+ (Setd8Δ/+) and EpoRCre+/-;Setd8+/+ (Setd8+/+) mice to anemic stress by retro-orbital bleeding. Setd8Δ/+ and Setd8+/+ mice had similar hematocrit after anemic stress (26.6 vs 29.4%; p=0.216), but the Setd8Δ/+ had an impaired ability to mount a stress response, with a lower MCV (43.0 vs 45.1 fL, p=0.003) and reticulocyte count (8.05 vs 2.14%, p=0.031) Consistent with the transcriptomic data, Setd8Δ/+ mice had higher levels of Fas transcript in splenic erythroblasts than Setd8+/+ controls. Together, these data suggest that high level Setd8 expression is important for normal erythroid maturation and gene expression, and for regulating the stress erythropoiesis response. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3795-3795
Author(s):  
Monika Belickova ◽  
Jaroslav Cermak ◽  
Alzbeta Vasikova ◽  
Eva Budinska

Abstract Abstract 3795 Poster Board III-731 Gene expression profiles of CD34+ cells were compared between a cohort of 51 patients with MDS or AML from MDS and 7 healthy controls. The patients were classified according to the WHO criteria as follows: 5q- syndrome (n=7), RA (n=3), RARS (n=2), RCMD (n=10), RAEB-1 (n=7), RAEB-2 (n=15), and AML with MLD (multilineage dysplasia) (n=7). HumanRef-8 v2 Expression Bead Chips (Illumina) were used to generate expression profiles of the samples for >22,000 transcripts. The raw data were normalized data with the R software, lumi package. Normalized data were filtered by detection p-value <0.01, resulting in total number of 9811genes. To identify differentially expressed genes we performed two parallel statistical hypothesis testings: Analysis of Variance (ANOVA) together with Tukey test and empirical bayesian thresholding correction for multiple testing problem; and Significance Analysis of Microarrays (SAM). The results were confirmed by real-time quantitative PCR for six genes (TaqMan Gene Expression Assays). Hierarchical clustering of significantly differentially expressed genes clearly separated patients and controls, 5q-syndrome and RAEB-1 as a separate entities confirming usefulness of WHO classification subgroups. The most up-regulated genes in all patients included HBG2, HBG1, CYBRD1, HSPA1B, ANGPT1, and MYC. We assume that expression changes in globin genes, both fetal and adult globins (HBG2, HBG1 and HBA1, HBB) may play role not only in dysregulation of erythropoiesis but also in the disease progression or leukemic transformation of MDS. Among the most down-regulated genes, 13 genes related to B-lymphopoiesis (e.g. POU2AF1, VPREB1, VPREB3, CD79A, EBF1, LEF1, BCL3, IRF8 & IRF4) were detected, suggesting the abnormal development of B-cell progenitors in all MDS patients. Some of these genes (e.g. VPREB3, LEF1) showed decreasing trend in expression level from early to advanced MDS with the lowest expression in AML with MLD. Patients with advanced MDS had significantly decreased expression of genes involved in in the mitotic cell cycle, DNA replication, and chromosome segregation compared to early MDS where these gene subsets were up-regulated. The DAVID database also identified de-regulation in the cell cycle pathway through its 7 genes (CDC25C, CDC7, CDC20, ORC1L, CCNB2, BUB1, & CCNA2). On the other hand, advanced MDS patients showed significant up-regulation of proto-oncogenes (BMI1, MERTK) and genes related to angiogenesis (ANGPT1), anti-apoptosis (VNN1). The results confirm on molecular basis that increased cell proliferation and resistance to apoptosis together with a loss of cell cycle control, damaged DNA repair and altered immune response may play an important role in the expansion of malignant clone in MDS patients. The study was supported by Grant NR-9235 obtained from the Ministry of Health, Czech Republic. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2407-2407
Author(s):  
Riccardo Bomben ◽  
Stefano Volinia ◽  
Stefania Gobessi ◽  
Daniela Marconi ◽  
Michele Dal Bo ◽  
...  

Abstract Abstract 2407 Introduction: The clinical course of chronic lymphocytic leukemia (CLL) is highly variable and prognosis is strongly associated with the mutational status of the IGHV genes. Recently, it has been observed that CLL cells expressing unmutated (UM) IGHV genes can be more efficiently induced to proliferate by stimulation of Toll-like Receptor 9 (TLR9) with unmethylated CpG oligonucleotides (CpG) than CLL cells expressing mutated (M) IGHV genes. MicroRNA (miRNAs) are 18- to 22-nucleotide-long RNA molecules that regulate gene expression and play a key role in several biological process including oncogenesis. Although the recognized pathogenetic relevance of miRNAs in CLL, their involvement in regulating activation/proliferation processes of CLL cells has still to be elucidated. Patients and Methods: Freshly-isolated negatively-selected CLL cells from 19 patients (9 UM and 10 M CLL) were stimulated with CpG or left unstimulated for 18 hours. MiRNA profiling and Gene expression profiling (GEP) were performed according to Agilent Technologies protocols. Bioinformatics analyses were performed integrating three different methods for supervised analysis (LIMMA algorithm, Agilent and Partek softwares). Results: The miRNA profile of CLL cells treated or not with CpG was separately evaluated in M and UM CLL. Consistent with the notion that M CLL cells are usually non-responsive to CpG stimulation, no miRNA was found to be differentially expressed between CpG-stimulated and unstimulated CLL cells belonging to this subgroup. In contrast, in UM CLL, as many as 28 miRNAs resulted differentially expressed, 24 up-regulated (miR-1260, miR-1274a, miR-1274b, miR-1280, miR-155, miR-155*, miR-17, miR-17*, miR-18a, miR-196a, miR-19b-1*, miR-20a, miR-20b, miR-221, miR-221*, miR-222, miR-29b-1*, miR-30b*, miR-30d*, miR-374b*, miR-720, miR-886-3p, miR-92a-1*, miR-939) and 4 down-regulated (miR-1226*, miR-125a-3p, miR-135a*, miR-150*) upon CpG stimulation. Data were confirmed by quantitative real time PCR. In order to identify the miRNAs actually involved in regulating activation/proliferation processes induced by TLR9 triggering, a concomitant GEP was performed comparing the same UM CLL cells exposed or not to CpG. Data analysis was carried out by taking advantage of the T-REX software that, by integrating four algorithms and six different target prediction programs, allows the identification of the regulated miRNAs on the basis of their repression activity on target mRNA. T-REX application selected four miRNAs whose mRNA targets resulted significantly down-regulated upon TLR9 triggering, namely miR-17, miR-20a, miR-20b and miR-93a. All these miRNAs belong to the miR-17~92 cluster family, known to be over-expressed in a variety of B-cell lymphomas, including diffuse large B-cell lymphoma, Burkitt lymphoma, follicular lymphoma and mantle cell lymphoma. Notably, three of these miRNAs and four additional miRNAs also belonging to the miR-17~92 cluster family (e.g. miR-17*, miR-18a, mir-19b-1* and mir-92a-1*) turned out to be among the 24 up-regulated miRNAs in CpG-stimulated UM CLL cells. In-silico analyses performed with the “Onto-Express” software, found that several differentially expressed genes were included in Gene Ontology (GO) categories related to regulation of cell proliferation, G1/S transition, apoptosis and NFkB signalling, in keeping with the typical proliferative response induced by CpG stimulation in UM CLL cells. The down-regulated genes included in these categories comprised CDKN1B/P27, CCNG2, NCOA3, E2F5, MAPK4, TRIM8, ZBTB4 and TP53INP1, all known target of miR-17~92 cluster family. Notably, the gene for the negative cell cycle regulator CDKN1B/P27 is also targeted by miR-221 and miR-222, two miRNAs both up-regulated in UM CLL cells upon CpG stimulation. Finally, transcripts for the proto-oncogene MYC also resulted over-expressed upon CpG stimulation. This observation may be relevant given the capacity of MYC to directly and positively regulate expression of miRNAs belonging to the miR-17~92 cluster family. Conclusion: Induction of the miR-17~92 family is a specific feature of UM CLL cells triggered through TLR9 and is associated with down-regulation of genes involved in cell cycle control and apoptosis regulation. MiRNAs belonging to the miR-17~92 family may represent promising novel targets for biological therapies of high risk CLL. Disclosures: No relevant conflicts of interest to declare.


1991 ◽  
Vol 11 (11) ◽  
pp. 5718-5726
Author(s):  
A Rowley ◽  
R A Singer ◽  
G C Johnston

The cell cycle of the budding yeast Saccharomyces cerevisiae has been investigated through the study of conditional cdc mutations that specifically affect cell cycle performance. Cells bearing the cdc68-1 mutation (J. A. Prendergast, L. E. Murray, A. Rowley, D. R. Carruthers, R. A. Singer, and G. C. Johnston, Genetics 124:81-90, 1990) are temperature sensitive for the performance of the G1 regulatory event, START. Here we describe the CDC68 gene and present evidence that the CDC68 gene product functions in transcription. CDC68 encodes a 1,035-amino-acid protein with a highly acidic and serine-rich carboxyl terminus. The abundance of transcripts from several unrelated genes is decreased in cdc68-1 mutant cells after transfer to the restrictive temperature, while at least one transcript, from the HSP82 gene, persists in an aberrant fashion. Thus, the cdc68-1 mutation has both positive and negative effects on gene expression. Our findings complement those of Malone et al. (E. A. Malone, C. D. Clark, A. Chiang, and F. Winston, Mol. Cell. Biol. 11:5710-5717, 1991), who have independently identified the CDC68 gene (as SPT16) as a transcriptional suppressor of delta-insertion mutations. Among transcripts that rapidly become depleted in cdc68-1 mutant cells are those of the G1 cyclin genes CLN1, CLN2, and CLN3/WHI1/DAF1, whose activity has been previously shown to be required for the performance of START. The decreased abundance of cyclin transcripts in cdc68-1 mutant cells, coupled with the suppression of cdc68-1-mediated START arrest by the CLN2-1 hyperactive allele of CLN2, shows that the CDC68 gene affects START through cyclin gene expression.


2002 ◽  
Vol 108 (4) ◽  
pp. 204-209 ◽  
Author(s):  
Koko Murakami ◽  
Hitoshi Kanno ◽  
Jakica Tancabelic ◽  
Hisaichi Fujii

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 549-549 ◽  
Author(s):  
Claudia Schoch ◽  
Alexander Kohlmann ◽  
Wolfgang Kern ◽  
Sylvia Merk ◽  
Wolfgang Hiddemann ◽  
...  

Abstract Deletions of the long arm of chromosome 5 occur either as the sole karyotype abnormality in MDS and AML or as part of a complex aberrant karyotype. It was the aim of this study to analyze the impact of the 5q deletion on the expression levels of genes located on chromosome 5q in AML and MDS. Therefore, gene expression analysis was performed in 344 AML and MDS cases using Affymetrix U133A+B oligonucleotide microarrays. The following subgroups were analyzed: AML with sole 5q deletion (n=7), AML with complex aberrant karyotype (n=83), MDS with sole 5q deletion (n=9), and MDS with complex aberrant karyotype (n=9). These were compared to 200 AML and 36 MDS with normal karyotype. In total, 1313 probe sets representing 603 genes cover sequences located on the long arm of chromosome 5. Overall a significant lower mean expression of all genes located on the long arm of chromosome 5 was observed in subgroups with 5q deletion in comparison to their respective control groups (for all comparisons, p&lt;0.05). 36 genes showed a significantly lower expression in all comparisons. These genes are involved in a variety of different biological processes such as signal transduction (CSNK1A1, DAMS), cell cycle regulation (HDAC3, PFDN1) and regulation of transcription (CNOT8). In addition we performed class prediction using support vector machines (SVM). In one approach all 6 different subgroups were analyzed as one class each. While AML and MDS with normal karyotype as well as AML with complex aberrant karyotype were correctly predicted with high accuracies (97%, 81%, and 92%, respectively) AML and MDS with 5q- sole and MDS with complex aberrant karyotype were frequently misclassified as AML with complex aberrant karyotype. In a second approach only two classes were defined: all cases with 5q deletion combined vs. all cases without 5q deletion. 102 out of 108 cases (94%) with 5q deletion were identified correctly supporting the fact that a distinct gene expression pattern is associated with 5q deletion in general. Performing SVM only with genes located on the long arm of chromosome 5 also resulted in a correct prediction of 92 of 108 (85%) stressing the importance of the expression of genes located on chromosome 5 for these AML and MDS subtypes. The top 100 differentially expressed probe sets between cases with and without 5q deletion represented 74 different annotated genes of which 23 are located on the long arm of chromosome 5. They are involved in a variety of different biological functions such as DNA repair (POLE, RAD21, RAD23B), regulation of transcription (ZNF75A, AF020591, MLLT3, HOXB6), protein biosynthesis (UPF2, TINP1, RPL12, RPL14, RPL15) cell cycle control (GMNN, CSPG6, PFDN1) and signal transduction (HINT1, STK24, APP, CAMLG). 10 of the top 74 genes associated with 5q deletion were involved in the CMYC-pathway with upregulation of RAD21, RAD23B, GMMN, CSPG6, APP, POLE STK24 and STAG2, and downregulation of ACTA2, and RPL12. Ten other genes out of the 74 top differentially expressed genes were involved in the TP53 pathway with upregulation of H1F0, PTPN11 and TAF2 and downregulation of DF, UBE2D2, EEF1A1, IGBP1, PPP2CA, EIF2S3, and NACA. In conclusion, loss of parts of the long arm of chromosome 5 leads to a lower expression of genes located on the long arm of chromosome 5. A specific pattern of functionally related genes was identified which shows a lower expression in AML and MDS subtypes with 5q deletion.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1367-1367
Author(s):  
Christine Gilling ◽  
Amit Mittal ◽  
Vincent Nganga ◽  
Vicky Palmer ◽  
Dennis D. Weisenburger ◽  
...  

Abstract Abstract 1367 Previously, we have shown that gene expression profiles (GEP) of CLL cells from lymph nodes (LN), bone marrow (BM), and peripheral blood (PB) are significantly different from each other. Among the major pathways associated with differential gene expression, a “tolerogenic signature” involved in host immune tolerance is significant in regulating CLL progression. The genes associated with the tolerogenic signature are significantly differentially expressed in patient LN-CLL compared to BM-CLL and PB-CLL, suggesting that LN-CLL cells induce this immune tolerance. From 83 differentially expressed genes identified by GEP that are associated with immune dysregulation, we selected eleven genes (CAV1, PTPN6, PKCb, ZWINT, IL2Ra, CBLC, CDC42, ZNF175, ZNF264, IL10, and HLA-G) for validation studies to determine whether these genes are also dysregulated in the Emu-TCL1 mouse model of CLL. The results demonstrate a trend of upregulation of these genes as determined by qRT-PCR in the LN-tumor microenvironment. To further evaluate the kinetics of selected gene expression during tumor progression, we determined the expression levels of Cav1, Ptpn6, and Pkcb at 12, 24, and 36 weeks of CLL development in the Em-TCL1 mouse model. We found that the expression of all three genes increased as a function of age, indicating a correlation of gene expression with disease progression. In addition, as CLL progressed in these mice there was a marked decrease in CD4+ and CD8+ T cells. The murine data were further validated using CLL cells from the same patients with indolent versus aggressive disease indicating a similar trend in expression as CLL progressed (n=4). Furthermore, patient data analyzed by Kaplan Meier analyses of the expression levels of the selected genes indicated a significant association between down-regulation of PTPN6 (p=0.031) and up-regulation of ZWINT (p<0.001) with clinical outcome as determined by a shorter time to treatment (p<0.05). Functional analysis by knockdown of CAV1 and PKCb in primary patient CLL cells determined by MTT assay showed a decrease in proliferation following knockdown of these genes (p<0.005). Protein-interaction modeling revealed regulation of CAV1 and PTPN6 by one another. Additionally, the PTPN6 protein regulates B cell receptor (BCR) signaling and subsequently the BCR regulates PKCb. Therefore, these data from both mice and humans with CLL, argue that an aggressive disease phenotype is paralleled by expression of genes associated with immune suppression. In particular, evidence presented here suggests, dysregulation of CAV1, PTPN6, ZWINT, and PKCb expression promotes CLL progression. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5023-5023
Author(s):  
Monika Belickova ◽  
Jaroslav Cermak ◽  
Jitka Vesela ◽  
Eliska Cechova ◽  
Zuzana Zemanova ◽  
...  

Abstract Abstract 5023 A direct effects of lenalidomide on gene expression in 5q- patients was studied using HumanRef-8 v2 Expression BeadChips (Illumina). Expression profiles of 6 patients (before treatment and at the time of the first erytroid response) and 6 healthy controls were investigated from CD14+ monocytes of peripheral blood. Differentially expressed genes were identified by Significance Analysis of Microarrays (SAM). Simultaneously, selected genes (TNF, JUN, IL1) were monitored in the course of treatment using Real-Time PCR with Taqman Gene Expression Assays. A comparison of gene expression levels before and during lenalidomide treatment revealed 97 differentially expressed genes (FC >2; p<0.05) related to following biological processes: immune response (16 genes), inflammatory response (11 genes), response to bacteria (8 genes), anti-apoptosis (7 genes), regulation of MAP kinase activity (5 genes), oxygen transport (4 genes), and regulation of cell proliferation (11 genes). An overexpression of a number of cytokines (e.g. TNF, IL8, IL1B, CCL3L, CXCL2, and TNFAIP3) was detected in patients before treatment, after lenalidomide administration expression of the majority of the up-regulated cytokine genes decreased to the control baseline level. Detected overproduction of the cytokines in 5q- syndrome may lead to an increased apoptosis of hematopoietic progenitor cells and together with excessive oxidative stress may contribute to the damage the hematopoietic niche. In the same manner, untreated patients showed suppressed expression of two genes (CXCR4, CRTAP) which play an important role in the stem cell niche. After treatment, we detected increased expression of these genes. Both the observations might explain favorable effects of lenalidomide on the bone marrow stroma defect seen in 5q- syndrome. On the other hand, a substantial increase of the ARPC1B gene (an activator and a substrate of Aurora A) expression was detected after lenalidomide treatment. Since overexpression of Aurora A leads to polyploidy and chromosomal instability, ARPC1B might play a role in the disease progression observed in some patients treated with lenalidomide. To conclude, described changes in genes expression may contribute to identification of the pathways affected by lenalidomide and to the explanation of some effects of this drug that have not been fully understood yet. Supported by grants NS/9634 MZCR, UHKT2005 00023736, MSM0021620808 and COST EUGESMA Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3020-3020
Author(s):  
Alicia Báez ◽  
Beatriz Martin-Antonio ◽  
Concepción Prats-Martín ◽  
Isabel Álvarez-Laderas ◽  
María Victoria Barbado ◽  
...  

Abstract Abstract 3020 Introduction: Hematopoietic progenitors cells (HPCs) used in allogenic transplantation (allo-HSCT) may have different biological properties depending on their source of origin: mobilized peripheral blood (PB), bone marrow (BM) or umbilical cord (UC), which may be reflected in miRNAs or gene expression. The identification of different patterns of expression could have clinical implications. The aim of this study was to determine differences in miRNAs and gene expression patterns in the different sources of HPCs used in allo-HSCT. Materials and Method: CD34 + cells were isolated by immunomagnetic separation and sorting from 5 healthy donors per type of source: UC, BM and PB mobilized with G-CSF. A pool of samples from PB not mobilized was used as reference group. We analyzed the expression of 375 miRNAs using TaqMan MicroRNA Arrays Human v2.0 (Applied Biosystems), and gene expression using Whole Human Genome Oligo microarray kit 4×44K (Agilent). The expression levels of genes and miRNAs were obtained by the 2-ΔΔCTmethod. From expression data hierarchical clustering was performed using the Euclidean distance. To identify genes and miRNAs differentially expressed between the different sources of HPCs statistical Kruskal Wallis test was applied. All analysis were performed using the Multiexperiment Viewer 4.7.1. The function of the miRNAs and genes of interest was determined from the various databases available online (TAM database, Gene Ontology and TargetScan Human). Results: Forty-two miRNAs differentially expressed between the different sources were identified. As compared to BM or UC, in mobilized PB most miRNAs were overexpressed, including the miRNA family of miR515, which is characteristic of embryonic stem cells. On the other hand, 47 genes differentially expressed between the different sources were identified. Interestingly, a similar pattern of expression was observed between movilized PB and UC as compared to BM. Interestingly, 13 of these genes are targets of the miRNAs also identified in this study, which suggests that their expression might be regulated by these miRNAs. Conclusion: There are significant differences in miRNAs and gene expression levels between the different sources of HPCs Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document