scholarly journals FOXO1 repression contributes to block of plasma cell differentiation in classical Hodgkin lymphoma

Blood ◽  
2014 ◽  
Vol 124 (20) ◽  
pp. 3118-3129 ◽  
Author(s):  
Marion J. Vogel ◽  
Linka Xie ◽  
Hanfeng Guan ◽  
Reuben M. Tooze ◽  
Thomas Maier ◽  
...  

Key Points FOXO1 directly activates PRDM1α, the master regulator of PC differentiation, and it enriches a PC signature in cHL cell lines. PRDM1α is a tumor suppressor in cHL.

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.


2005 ◽  
Vol 23 (3-4) ◽  
pp. 127-132 ◽  
Author(s):  
Maike Buettner ◽  
Axel Greiner ◽  
Athanasia Avramidou ◽  
Hans-Martin Jäck ◽  
Gerald Niedobitek

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.


2008 ◽  
Vol 38 (8) ◽  
pp. 2316-2324 ◽  
Author(s):  
Francisco Mora-López ◽  
Nuria Pedreño-Horrillo ◽  
Luis Delgado-Pérez ◽  
José A. Brieva ◽  
Antonio Campos-Caro

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 148-148
Author(s):  
Jonathan Mandelbaum ◽  
Qiong Shen ◽  
Hongyan Tang ◽  
Tongwei Mo ◽  
Barbara Malynn ◽  
...  

Abstract Abstract 148 Diffuse large B-cell lymphoma (DLBCL), the most common type of non-Hodgkin lymphoma, is a heterogeneous disease comprising multiple biologically and clinically distinct subgroups, including germinal center B cell-like (GCB) and activated B cell-like (ABC) DLBCL. Numerous genetic alterations segregate with ABC-DLBCL, namely translocations of the BCL6 proto-oncogene, BLIMP1 inactivation and constitutive NF-κB activation. We recently reported that A20, a negative regulator of NF-κB signaling, is biallelically inactivated by mutations and deletions in one-third of ABC-DLBCL (Compagno et al, Nature, 2009), indicating a tumor suppressor role in this disease. Notably, A20 inactivation is commonly associated with chromosomal translocations deregulating BCL6 (n=11/20 DLBCL cases). Furthermore, the two genes are linked in the same pathway, where NF-κB induced activation of IRF4 leads to BCL6 down-regulation (Saito et al. Cancer Cell 2007) and consequent release of the BCL6 target BLIMP1, a master regulator of plasma cell differentiation. These observations suggest that A20 inactivation and BCL6 translocations cooperate in DLBCL pathogenesis. In order to examine the individual and combined contribution of these two lesions in vivo, we have generated an A20 conditional knockout allele in which a loxP-flanked exon 3 of the A20 gene can be deleted upon Cre-mediated recombination. The resulting mice were crossed with both a Cγ1-Cre deletor strain, which expresses the Cre recombinase in germinal centre (GC) B cells, and the lymphoma-prone Iμ HABCL6 mouse model (Cattoretti et al., Cancer Cell, 2005), which mimics a BCL6 translocation to the immunoglobulin heavy chain locus. When analyzed at 3 months of age, GC B-cell conditional heterozygous (A20Cγ1HET) and homozygous (A20Cγ1KO) A20 knockout mice showed a significant increase in the B220dimCD138+ plasma cell population (0.6% and 0.5%, respectively, versus 0.3% for wild-type littermates) and a corresponding 2-fold increase in IgG1 serum immunoglobulin levels after immunization with sheep red blood cells. Furthermore, A20 knockout splenic B cells had increased proliferative capacity and survival after stimulation ex vivo with lipopolysaccharides, B-cell receptor cross-linking or CD40 activation, consistent with enhanced NF-κB activity in these cells. Interestingly, the increase in plasma cells was not observed in compound Iμ HABCL6/ A20Cγ1HET and Iμ HABCL6/ A20Cγ1KO animals, presumably due to the known role of BCL6 in blocking plasma cell differentiation (Tunyaplin et al., J. of Immunol., 2004). In contrast, these animals displayed a marked increase in the B220+PNAhi GC B cell compartment, as compared to both A20 knockout and Iμ HABCL6 mice (2.5% and 3.9% respectively, versus 1.9% for wild-type littermates). Overall, these findings document that A20 acts as a negative regulator of B cell proliferation and survival as well as of plasma cell differentiation in vivo, and support a model by which loss of A20 synergizes with BCL6 deregulation to promote the expansion of GC B cells while preventing terminal differentiation. Long-term follow-up of these cohorts will provide critical information on the role of A20 as a tumor suppressor gene in vivo and on its cooperative activity with BCL6 deregulation in the pathogenesis of DLBCL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 129 (11) ◽  
pp. 1480-1490 ◽  
Author(s):  
Malena Zahn ◽  
Ralf Marienfeld ◽  
Ingo Melzner ◽  
Janine Heinrich ◽  
Benjamin Renner ◽  
...  

Key Points A novel PTP1B variant, PTP1BΔ6, is expressed in cHL cell lines and tumor samples. PTP1BΔ6 augments JAK/STAT activity, cell proliferation, and survival in cHL cell lines.


Blood ◽  
2015 ◽  
Vol 126 (18) ◽  
pp. 2098-2109 ◽  
Author(s):  
Martine van Keimpema ◽  
Leonie J. Grüneberg ◽  
Michal Mokry ◽  
Ruben van Boxtel ◽  
Menno C. van Zelm ◽  
...  

Key Points Aberrant expression of FOXP1 in human MBCs represses their ability to differentiate into PCs. Human IgG+ MBCs combine lower FOXP1 expression with a higher propensity to differentiate as compared with IgM+ MBCs.


Blood ◽  
2013 ◽  
Vol 121 (6) ◽  
pp. 962-970 ◽  
Author(s):  
Hiromichi Yuki ◽  
Shikiko Ueno ◽  
Hiro Tatetsu ◽  
Hiroaki Niiro ◽  
Tadafumi Iino ◽  
...  

Key PointsPU.1 is a potent tumor suppressor in cHL cells and the induction of PU.1 is a possible therapeutic option for patients with cHL.


Blood ◽  
2018 ◽  
Vol 131 (14) ◽  
pp. 1556-1567 ◽  
Author(s):  
Clarissa D. Osswald ◽  
Linka Xie ◽  
Hanfeng Guan ◽  
Franziska Herrmann ◽  
Sarah M. Pick ◽  
...  

Key Points FOXO3A expression indicates an abortive PC differentiation state in cHL. Tight regulation of FOXO3A is essential for the cHL oncogenic program.


Sign in / Sign up

Export Citation Format

Share Document