Exome Sequencing and Linkage Analysis Implicates Two Candidate Genes On Chromosome 3p in Familial Hodgkin Lymphoma

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 53-53 ◽  
Author(s):  
Alastair Lawrie ◽  
Timothee Cezard ◽  
Dominic J Culligan ◽  
Mark A Vickers

Abstract Abstract 53 Background It is well recognised that a genetic component exists in classical Hodgkin lymphoma. Higher rates are seen in first-degree relatives of affected individuals, with high concordance observed in monozygotic compared to dizygotic twins. Numerous associations with HLA alleles have been demonstrated in both Epstein-Barr virus-positive and negative forms of the disease and several non-HLA genes have now been implicated. However, these genetic factors are insufficient to account for all the observed inherited risk. Methods We report a family from North-East Scotland containing five related individuals with classical Hodgkin lymphoma. DNA was extracted from either whole peripheral blood or saliva. Affected individuals were genotyped using the Affymetrix 500Kb SNP array. Genome-wide linkage analysis was performed using easyLINKAGE plus. Targeted exome capture was performed on four affected individuals using the Agilent SureSelect 50Mb kit. Exome sequencing was performed on the Illumina HiSeq2000 to produce 101bp paired-end reads. Following quality control, raw fastq reads were aligned to the human genome (GRCh37) using bwa. SNP/indel calls were performed using Samtools. Variants identified were filtered by quality to include only those with a read coverage of ≥ 20 and phred score of ≥ 30. Given the high penetrance in this family, we hypothesised that a causative genetic variant would not have been described before so eliminated all SNPs present in dbSNP (version 132) or the 1000 genomes project. Finally, only SNPs affecting protein structure were considered further. SIFT and Polyphen-2 were used to predict impact of genetic variation upon protein structure/function. Results Karyotypic analysis was performed on one individual at diagnosis. This was normal, suggesting that a consitutional chromosomal disorder is not responsible for classical Hodgkin lymphoma in this family. The two regions exhibiting strongest linkage with disease were observed on chromosome 3p (LOD scores 1.5 & 1.4). Exome sequencing revealed seven novel, non-synonymous, heterozygous SNPs present in all four analysed individuals. Three were considered biologically plausible (FAM107A:A89S, ALS2CL:L440V, IGSF3:E414G) on the basis of known function and pattern of expression. Two of these three genes (FAM107A and ALS2CL) are on chromosome 3p, in regions similar to those identified in the linkage analysis, and have been implicated as candidate tumor suppressor genes in other malignancies. All three variants were located in evolutionary conserved regions. However, only the mutations in FAM107A and IGSF3 were predicted to disrupt protein function. Discussion Two genes on chromosome 3p represent candidate loci for causing familial classical Hodgkin lymphoma. FAM107A protein is downregulated in several malignant cell lines and primary tumor cells. Overexpression can result in suppression of tumor growth. A study of head and neck carcinoma identified frequent missense mutations and/or loss of heterozygosity at ALS2CL, suggesting that this gene can contribute to tumorigenesis. We are evaluating these genes further by assessing gene expression and protein levels in both Hodgkin cell lines and primary Reed-Sternberg cells. Finally, we intend to evaluate the presence of these candidate genetic variants in other, unaffected members of this family and sporadic cases. Disclosures: No relevant conflicts of interest to declare.

Author(s):  
A Ustaszewski ◽  
J Paczkowska ◽  
J Janiszewska ◽  
S Hartmann ◽  
J Bein ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (10) ◽  
pp. 3326-3334 ◽  
Author(s):  
Alexey Ushmorov ◽  
Olga Ritz ◽  
Michael Hummel ◽  
Frank Leithäuser ◽  
Peter Möller ◽  
...  

Abstract Immunoglobulin production is impaired in Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) in spite of functional clonal rearrangements. The presence of “crippling” mutations in coding and regulatory regions, as well as down-regulation of B-cell-specific transcription factors, has been suggested as a potential reason for the lack of immunoglobulin (Ig) chain gene transcription. We have investigated the impact of epigenetic silencing in suppressing Ig heavy (H)-chain expression. Chromatin immunoprecipitation (ChIP) was used to analyze transcription factor binding to octamer motifs present in the IgH regulatory regions. Transcription factors were bound to these motifs in control cell lines, however, they were absent in the cHL-derived cell lines KMH2, L1236, and L428. Ectopic expression of octamer-binding transcription factor (Oct2) and/or B-cell Oct binding protein/Oct-binding factor (BOB.1/OBF.1) did not result in any measurable binding to these sites. Increased histone 3 Lysine 9 (H3-K9) methylation was observed in the promoter region of the IgH locus in L428 and L1236 cells. This is a typical feature of heterochromatic, transcriptionally silent regions. Treatment of cHL-derived cell lines with the DNA demethylating agent 5-aza-2′-deoxycytidine (5-aza-dC) partially reactivated IgH transcription and affected chromatin modifications. Our results suggest an important role of epigenetic silencing in the inhibition of IgH transcription in HRS cells. (Blood. 2004;104:3326-3334)


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 19-19 ◽  
Author(s):  
Marc A. Weniger ◽  
Ingo Melzner ◽  
Christiane K. Menz ◽  
Silke Wegener ◽  
Alexandra J. Bucur ◽  
...  

Abstract The suppressors of cytokine signaling (SOCS) are critically involved in the regulation of cellular proliferation, survival, and apoptosis via cytokine-induced JAK/STAT signaling. SOCS-1 silencing by aberrant DNA methylation contributes to oncogenesis in various B-cell neoplasias and carcinomas. Recently, we showed an alternative loss of SOCS-1 function due to deleterious SOCS-1 mutations in a major subset of primary mediastinal B-cell lymphoma (PMBL) and in the PMBL line MedB-1, and a biallelic SOCS-1 deletion in PMBL line Karpas1106P (BLOOD, 105, 2535–42, 2005). For both cell lines our previous data demonstrated retarded JAK2 degradation and sustained phospho-JAK2 action leading to enhanced DNA binding of phospho-STAT5. Here we analysed SOCS-1 in laser-microdissected Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL). We detected SOCS-1 mutations in HRS cells of eight of 19 cHL samples and in three of five Hodgkin lymphoma (HL)-derived cell lines by sequencing analysis. Moreover, we found a significant association between mutated SOCS-1 of isolated HRS cells and nuclear phospho-STAT5 accumulation in HRS cells of cHL tumor tissue (p<0.01). Collectively, these findings support the concept that PMBL and cHL share many overlapping features, and that defective tumor suppressor gene SOCS-1 triggers an oncogenic pathway operative in both lymphomas.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2628-2628
Author(s):  
Hiromichi Yuki ◽  
Shikiko Ueno ◽  
Hiroaki Niiro ◽  
Hiro Tatetsu ◽  
Hiroyuki Hata ◽  
...  

Abstract Abstract 2628 PU.1 is an Ets family transcription factor, which is essential for differentiation of both myeloid and lymphoid lineage cells. We have previously shown that PU.1 is down-regulated in various myeloma cell lines and myeloma cells from a subset of myeloma patients. In such cell lines, the promoter and the upstream regulatory element (URE) located in 17 kb 5'-upstream of the PU.1 gene are highly methylated. Furthermore, conditionally expressed PU.1 induces both cell growth arrest and apoptosis in PU.1-low to -negative myeloma cell lines, U266 and KMS12PE. Therefore, we concluded that the down-regulation of PU.1 is necessary for myeloma cell growth. In another B cell malignancy, classical Hodgkin lymphoma, it has been reported that PU.1 is also down-regulated through methylation of its promoter. To evaluate whether down-regulation of PU.1 is essential for growth of classical Hodgkin lymphoma cells, we conditionally expressed PU.1 in two classical Hodgkin lymphoma cell lines, L428 and KMH2, using the tet-off system (designated as L428tetPU.1 and KMH2tetPU.1 cells, respectively). Up-regulation of PU.1 by tetracycline removal induced complete growth arrest in L428tetPU.1 and KMH2tetPU.1 cells. Annexin V staining revealed that up-regulation of PU.1 induced apoptosis in both cell lines. Furthermore, BrdU staining analysis revealed that PU.1 induced G0/G1 arrest in those cells. L428tetPU.1 and KMH2tetPU.1 cells expressing PU.1 showed morphological changes that included the enlargement cytosol and the appearance of various sizes of vacuoles. We next injected L428tetPU.1 and KMH2tetPU.1 cells to immunodeficiency mice (Rag2−/− Jak3−/− bulb/c) subcutaneously. Tumor formation was observed in all those mice with continuous administration of tetracycline (0.5 g/l) in the drinking water. After enlargement of tumor to 1–2 cm diameter, we removed tetracycline in half of the mice. Tetracyclin withdrawal resulted in tumor regression or stable disease, whereas all the mice continuously receiving tetracycline had continuous tumor growth and finally died. These data strongly suggest that PU.1 induced growth arrest and apoptosis of classical Hodgkin lymphoma cells both in vitro and vivo. We next performed DNA microarray analysis to compare gene expression levels of L428tetPU.1 cells before and after PU.1 expression to elucidate the mechanisms of growth arrest and apoptosis induced by PU.1. Among genes related to cell cycle and apoptosis, p21 (CDKN1A) was highly up-regulated in L428tetPU.1 cells after PU.1 induction, and this was also confirmed by mRNA and protein levels. Finally, to clarify the role of p21 up-regulation by PU.1, we stably introduced p21 siRNA in L428tetPU.1 cells. Such stably expressed p21 siRNA rescued L428tetPU.1 cells from growth arrest induced by PU.1, suggesting that the growth arrest in L428tetPU.1 cells by PU.1 should be at least partially dependent on p21 up-regulation. These data suggested that up-regulation of PU.1 by demethylation agents and/or HDAC inhibitors might serve as a possible treatment modality for classical Hodgkin lymphoma. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3645-3645
Author(s):  
Paul Greaves ◽  
Sameena Iqbal ◽  
David C Taussig ◽  
John G. Gribben

Abstract Abstract 3645 Introduction: The bulk of the tumour infiltrate in classical Hodgkin lymphoma (CHL) is composed of immune cells, predominantly CD4+ T cells, with the malignant Hodgkin Reed Sternberg cell (HRS) representing <1% of cells. The lymphoid microenvironment has been described as anergic and hypoproliferative with suppressive properties (Marshall et al. Blood 2004 103:1755–62) but the functional significance of this is unclear. This study set out to examine the proliferative capacity and phenotype of T cells derived from CHL-diagnostic lymph node tissue taken at diagnosis. Method: Frozen single cell suspensions (SCS) from 6 patients were selected from the tissue bank of our Institute. T cell growth-augmenting and/or Th2 polarising cytokines were added in various combinations (IL2, IL4 only, IL2+4 or no added cytokine) to SCS-derived cells in 96 well plates at 0.3 × 106 cells per well in 200mcl of optimized lymphocyte culture media. No CD4+ enrichment step was carried out: all recovered cells were plated at baseline to maintain potential interactions between CD4+ cells and other cells, and no mitogen or T-cell receptor-stimulating or costimulating agents were added at any point. As controls, SCS derived from normal tonsil, and ÔreactiveÕ lymph nodes (n=4) (confirmed by histological report at the time of diagnosis) were also plated. Plates were examined daily for cell/colony morphology to estimate growth and split with fresh media and cytokines once every 7 days, with estimated proliferation (by haemocytometry) plotted. Cultures were assessed at baseline, 10 days, 28 days, 50 days and 100 days. Results: Proliferation, based on formation of discrete colonies and blastoid cell morphology, occurred in the majority of wells by day 7 in all CHL-derived cultures, and in a minority of wells, and to a lesser extent in all control cultures. CHL-derived T cells from one patient continue to expand after 200 days, doubling every 3–5 days, while the other 5 continue after 50–100 days. In contrast, no tonsil or reactive node-derived T cells survived beyond 50 days and none showed a net expansion in cell numbers. Growth was superior in the IL2+4 and IL2-only conditions, with no growth in the media-only or IL4-only conditions. The most favorable condition was with the addition of IL2+4. By day 21 a net increase in CHL-derived T cells was apparent, but not in any control T cell populations (Figure). At baseline, composition of the CHL-derived cells revealed a majority of CD3+ cells as expected, of which 60–80% were CD4+ and the remainder CD8+. By D21 the CD4+ component had outgrown all other cells in the CHL-derived cultures, being CD3+CD4+CD45RO+ consistent with antigen-experienced T helper cells, while all tonsil and reactive node-derived cells were CD8+CDRO+. Markers of central memory (CCR7 & CD62-L), Th2 (CCR4 and IL4), Treg (FOXP3 and CD25) and anergy (CD57) were absent after expansion, while markers of activation were upregulated (CD28, CD27, CD69, CD40L, CD30 & CD95). This phenotype persisted in the ongoing T cell lines. Conclusions: The CD4+ compartment of the CHL microenvironment contains a primed subset of cells capable of massive expansion without further mitogenic stimulation and of generating cytokine-dependent continuous cell lines with an antigen-experienced, activated phenotype. This challenges the assumption of T cell anergy and hypoproliferation in the tissue microenvironment of CHL. We are currently assessing the function and anti-tumor specific or tumor supportive nature of these T cells. Disclosures: Gribben: Roche: Honoraria; Celgene: Honoraria; GSK: Honoraria; Mundipharma: Honoraria; Gilead: Honoraria; Pharmacyclics: Honoraria.


2016 ◽  
Vol 12 (1) ◽  
pp. 219-232 ◽  
Author(s):  
D. Vergara ◽  
P. Simeone ◽  
S. De Matteis ◽  
S. Carloni ◽  
P. Lanuti ◽  
...  

Classical Hodgkin lymphoma models of T- and B-cell derivation show significant differences in their protein expression profiles.


2021 ◽  
Author(s):  
Felicia Gomez ◽  
Matthew Mosior ◽  
Joshua McMichael ◽  
Zachary L Skidmore ◽  
Eric J Duncavage ◽  
...  

The malignant Hodgkin and Reed Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) are scarce in affected lymph nodes, creating a challenge to detect driver somatic mutations. As an alternative to cell purification techniques, we hypothesized that ultra-deep exome sequencing would allow genomic study of HRS cells, thereby streamlining analysis and avoiding technical pitfalls. To test this, 31 cHL tumor/normal pairs were exome sequenced to ~1000x median depth of coverage. An orthogonal error-corrected sequencing approach verified >95% of the discovered mutations. We identified mutations in genes novel to cHL including: CDH5 and PCDH7; novel mutations in IL4R, and a novel pattern of recurrent mutations in pathways regulating Hippo signaling. This study provides proof-of-principle that ultra-deep exome sequencing can be utilized to identify recurrent mutations in HRS cells, allowing for the analysis for clinically relevant genomic variants in large cohorts of cHL patients.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1448-1448
Author(s):  
Ta-Chih Liu ◽  
Yuan-Shiang Kao ◽  
Rachael Demuth ◽  
Nina Mathews ◽  
Carmen Espinoza ◽  
...  

Abstract The paucity of Hodgkin cell/Reed-Sternberg cell in classical Hodgkin Lymphoma (HL) represents a general problem for molecular and cytogenetic studies. The established HL cell lines could be used for such studies. However, only about 10 cell lines have been established and all of them were isolated from patients in late stage of illness when the tumor had recurred. Only one of these cell lines is known to be positive for Epstein- Barr virus antigen. In addition, the pattern of chromosomal aberration in these cell lines is highly complex. Therefore, it is necessary to have a cell line established from the early stage of disease, without prior treatment and with less chromosomal aberration for research. A sample of left axillary lymph node from a 27-year-old male with early stage of HL, and without prior chemotherapy, was cultured in RPMI 1640 media supplemented with fetal calf serum for routine cytogenetic study. The culture, passed weekly, now in its 60st week, 57th passes is growing autonomously without supplement of any growth factor. The original lymph node biopsy showed a classical Hodgkin lymphoma, mixed cellularity (WHO Classification) and the Hodgkin cells/Reed-Sternberg cells were positive for CD30, 4+(100%), CD20, 2+(33%) and negative for CD3, CD15, CD43, ALK-1 antigen and epithelial membrane antigen. EBV nuclear antigen 1 DNA and RNA are detected by PCR method. The cell line is positive for CD30, CD20, and Epstein-Barr virus (EBV) latent membrane protein, but negative for CD3, CD79a, and EBV early antigen. By florescent insitu hybridization the cell line is negative for p53 deletion, ALK gene rearrangement, MLL gene deletion, and t(12;21). A ploidy analysis by flow cytometry shows 80.22% diploid, and 19.78% hyperploids. The cells have doubling time of 30 to 36 hours. The initial karyotypes were: 45~46, XY, i(3)(q10), der(6)t(3;6)(p11;q22), t(6;13) (p21;q32), del(7)(q32), i(14) (q10)[cp3]/46, XY, del(3)(p10), der(6)t(3;6), der(6)t(6;13), del(7)(q32), add(10)(p13), der(13)add(13)(p11.1)t(6;13), i(14)(q10)[3]/46, XY[16]. Metaphase preparations from the cell line showed 46, XY, absence of the above mentioned chromosomal abnormalities, but 97% (1422/1466 cells) of cells were diploid; 2.5% (36/1466 cells) of cells were tetraploid/near tetraploid, and 0.5% (8/1466 cells) of cells showed endore-duplication. Chromosomal comparative genomic hybridization of the cell line showed microdeletion on chromosome 5q34 and 13q22~31 region, and gain on 12q12.1. Single nucleotide polymorphism array showed no abnormalities. This newly established cell line is unique in: it is the second cell line known to be positive for EBV antigen, it shows no complex chromosomal aberration by conventional karyotyping or molecular genotyping, and since the cell line showed mostly in diploid, and only 3% of the cells are tetraploid/near tetraploid and endoreduplication by conventional cytogenetic method, the cell line is ideal for the study of formation of hyperploid cells (i.e. Reed-Sternberg cells) from diploid cells.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3794-3794
Author(s):  
Marion J. Vogel ◽  
Linka Xie ◽  
Hanfeng Guan ◽  
Clarissa D. Weitzer ◽  
Frank Leithäuser ◽  
...  

Abstract In contrast to other B cell lymphomas, Hodgkin and Reed-Sternberg (HRS) cells, the malignant cells of classical Hodgkin lymphoma (cHL), have mainly lost their B cell identity. Recently, we reported that the transcription factor FOXO1, indispensable for B cell development and differentiation, is downregulated in HRS cells and expression of FOXO1 results in growth arrest and apoptosis in cHL cell lines. To find molecular targets of FOXO1 we performed gene expression profiling of 5 cHL cell lines expressing constitutively active FOXO1 fused to ligand binding domain of estrogen receptor (FOXO1(A3)ER). We found that FOXO1 activation led to downregulation of genes, which are normally upregulated in cHL, such as CD30/TNFRSF8 and the proto-oncogene MYC. On the other hand, FOXO1 activated expression of germinal center-specific genes including BCL6, AICDA, BACH2, and GCET2. The most surprising finding was induction of BLIMP-1/PRDM1, the master regulator of plasma cell differentiation and tumor suppressor in activated B cell-like diffuse large B cell lymphoma (ABC-DLBCL). A positive correlation of FOXO1 and BLIMP-1 expression in microdissected HRS cells further indicated a role for FOXO1 in BLIMP-1 regulation. Of note, HRS cells do not undergo plasma cell differentiation despite constitutive expression of NF-κB, IRF4, and STAT3, known inducers of BLIMP-1. However, BLIMP-1 levels remain low in HRS cells thereby preventing terminal differentiation. The mechanisms leading to BLIMP-1 repression in cHL are poorly understood. We found that in most cHL cell lines, FOXO1 specifically upregulated BLIMP-1α representing the full-length isoform of BLIMP-1. In addition, luciferase reporter assay showed that FOXO1 activates the BLIMP-1α promoter in the cHL cell line L428. Overexpression of BLIMP-1α using a lentiviral vector strongly inhibited growth of cHL cell lines. In line with the fact that BLIMP-1 and MYC form a negative feedback loop, we found that ectopic BLIMP-1α expression resulted in downregulation of MYC protein levels, which most likely contributes to the anti-tumor effect of BLIMP-1α. On the other hand, MYC inhibition in cHL cell lines led to BLIMP-1 upregulation. Thus, our data indicate that FOXO1, BLIMP-1 and MYC constitute a negative feed-forward regulatory loop, which is controlled by FOXO1. Searching for additional mechanisms of BLIMP-1α downregulation in cHL we found that BLIMP-1α promoter was hypermethylated in two cHL cell lines but not in microdissected HRS cells. This indicates that hypermethylation is not critical for BLIMP-1α repression. Interestingly, the functionally impaired BLIMP-1 variant BLIMP-1β, which is normally not detected in normal B cells, was upregulated in cHL cell lines. BLIMP-1β is also expressed in ABC-DLBCL and has been suggested to block BLIMP-1α in a dominant-negative manner. We speculate that BLIMP-1β attenuates function of residual BLIMP-1α in cHL. However, further investigation is required. Taken together, we show for the first time that FOXO1 induces expression of BLIMP-1. We identified BLIMP-1α as a tumor suppressor which downregulates expression of the proto-oncogene MYC in cHL. Therefore, FOXO1 might exert its tumor suppressor function at least in part by upregulation of BLIMP-1α. Further work on BLIMP-1α regulation by FOXO1 and investigation of a potential role of FOXO1 in plasma cell differentiation is warranted. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 629-629 ◽  
Author(s):  
Alfons Navarro ◽  
Anna Gaya ◽  
Anna Cordeiro ◽  
Blanca Gonzalez-Farre ◽  
Marina Díaz-Beyá ◽  
...  

Abstract Introduction The behavior of classical Hodgkin lymphoma (cHL) is determined by both the intrinsic features of the Hodgkin/Reed Sternberg (HRS) cells and the characteristics of the microenvironment, making the analysis of entire lymph nodes an effective approach to understanding the disease. One of the genetic lesions most frequently found in HRS cells involves members of the JAK/STAT signaling pathway. However, few studies have identified microRNAs (miRNAs) that target JAK2 in cHL. In a previous study, our group identified miR-135a as a JAK2 regulator and showed the prognostic impact of this miRNA in a small set (n=89) of cHL patients (Navarro A. et al. Blood 2009). In the present study, we have examined novel miRNAs targeting JAK2 and evaluated the prognostic impact of their expression levels in 170 lymph nodes of cHL patients. Methods TargetScan and miRbase were used to identify JAK2-related miRNAs. The conserved miRNAs with higher scores were selected and further validated by Renilla-Luciferase assay and Western Blot. We performed a Renilla-Luciferase assay with a modified expression vector psiChecK-2 containing the complete 3’UTR region of JAK2 cloned in the 3’UTR region of Renilla luciferase gene. For Renilla-Luciferase assay, 100nM of the pre-miRNAs of interest or pre-miR negative control were transfected in the HDMYZ cHL cell line together with 1μM of modified psicheck2 vector using Lipofectamine 2000, and luminescence was read at 24 hours. Further validation of the miRNA target was performed by Western Blot in three cHL cell lines (L428, L-1236 and HDMYZ). The expression levels of the miRNAs identified as targeting JAK2 were then assessed in lymph nodes from 170 cHL patients (median age, 33 years; male, 51%; EBV, 43.8%; HIV, 12%) diagnosed and treated in a single institution. Moreover, 15 reactive lymph nodes (RLN) were used as normal controls. RNA was purified from formalin-fixed paraffin-embedded lymph nodes using RecoverAll Total Nucleic Acid Isolation kit. The miRNA expression was analyzed using TaqMan microRNA assays. Statistical analyses were performed using R v2.13 and PASW Statistics 18. Results The bioinformatic analysis identified seven microRNAs as possible regulators of JAK2 (miR-34a, miR-101, miR-135a, miR-204, miR-216a, miR-216b and miR-375). The Renilla-Luciferase assay confirmed that in addition to miR-135a, miR-101 (61%; p=0.01), miR-204 (46%; p=0.003) and miR-216a (64.8%; p=0.02) also targeted JAK2. These findings were confirmed by Western Blot analysis of JAK2 levels after transfection with pre-miRNAs (median % of protein reduction in the 3 cell lines of 21.1%, 46.7% and 24%, respectively). The analysis of these miRNAs in RLN showed that miR-101, miR-135a and miR-204 were significantly downregulated in lymph nodes of cHL patients (p<0.001, p<0.001 and p=0.02 respectively) in comparison with RLN used as control. In contrast, miR-216 did not show significant differences in comparison with RLN. Interestingly, the analysis of the expression levels of these 4 miRNA in the patient lymph nodes had an impact on prognosis. First, low expression levels of miR-135a was associated with lower rate of complete response to first line treatment (76% vs 89%, p=0.012) and a trend was observed for miR-216(p=0.077). Secondly, patients with low expression levels of miR-101 or miR-204 had shorter disease-free survival (DFS) (151 vs 109 months, p=0.020; and 149.2 vs 105.5 months, p=0.055). Finally, patients with low levels of miR-135a, miR-204 or miR-216 had shorter overall survival (OS) (158 vs 136 months; p=0.039; and 157.5 vs 110 months, p=0.025; and 165 vs 140 moths, p=0.011 respectively). Since all four miRNAs showed prognostic impact (DFS, OS or treatment response), we decide to analyze the prognostic impact of the combination of the 4 miRNAs. Patients were then classified in two subgroups according to the expression levels of all four miRNAs. Patients with low expression of more than 2 miRNAs had shorter OS (163 vs 132 months; p=0.012). The multivariate analysis identified the combination of the four miRNAs as an independent prognostic marker of OS (OR, 7.6; 95% CI, 1.1-57.2; p=0.048) together with Age≥45years (OR, 5.9; 95% CI, 2.4-14.4; p<0.001) and B symptoms (OR, 2.6; 95% CI, 1.09-6.3; p=0.03). Conclusions MiR-101, miR-135a, miR-204 and miR-216 regulate JAK2 and are independent prognostic factors in cHL. Acknowledgments SDCSD of School of Medicine of UB. AC is an APIF-UB fellow. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document