Evidence of abortive plasma cell differentiation in Hodgkin and Reed-Sternberg cells of classical Hodgkin lymphoma

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


Cell Reports ◽  
2016 ◽  
Vol 17 (1) ◽  
pp. 193-205 ◽  
Author(s):  
Vincent Biajoux ◽  
Jessica Natt ◽  
Christelle Freitas ◽  
Nagham Alouche ◽  
Antoine Sacquin ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (11) ◽  
pp. 1895-1898 ◽  
Author(s):  
Christelle Vincent-Fabert ◽  
Remi Fiancette ◽  
Eric Pinaud ◽  
Véronique Truffinet ◽  
Nadine Cogné ◽  
...  

Abstract The immunoglobulin heavy chain locus (IgH) undergoes multiple changes along B-cell differentiation. In progenitor B cells, V(D)J assembly allows expression of μ heavy chains. In mature B cells, class switch recombination may replace the expressed constant (C)μ gene with a downstream CH gene. Finally, plasma cell differentiation strongly boosts IgH transcription. How the multiple IgH transcriptional enhancers tune these changes is unclear. Here we demonstrate that deletion of the whole IgH 3′ regulatory region (3′RR) allows normal maturation until the stage of IgM/IgD expressing lymphocytes, but nearly abrogates class switch recombination to all CH genes. Although plasma cell numbers are unaffected, we reveal the role of the 3′RR into the transcriptional burst normally associated with plasma cell differentiation. Our study shows that transcriptional changes and recombinations occurring after antigen-encounter appear mainly controlled by the 3′RR working as a single functional unit.


Blood ◽  
2011 ◽  
Vol 117 (22) ◽  
pp. 5907-5917 ◽  
Author(s):  
Katerina Vrzalikova ◽  
Martina Vockerodt ◽  
Sarah Leonard ◽  
Andrew Bell ◽  
Wenbin Wei ◽  
...  

AbstractAn important pathogenic event in Epstein-Barr virus (EBV)-associated lymphomas is the suppression of virus replication, which would otherwise lead to cell death. Because virus replication in B cells is intimately linked to their differentiation toward plasma cells, we asked whether the physiologic signals that drive normal B-cell differentiation are absent in EBV-transformed cells. We focused on BLIMP1α, a transcription factor that is required for plasma cell differentiation and that is inactivated in diffuse large B-cell lymphomas. We show that BLIMP1α expression is down-regulated after EBV infection of primary germinal center B cells and that the EBV oncogene, latent membrane protein-1 (LMP-1), is alone capable of inducing this down-regulation in these cells. Furthermore, the down-regulation of BLIMP1α by LMP-1 was accompanied by a partial disruption of the BLIMP1α transcriptional program, including the aberrant induction of MYC, the repression of which is required for terminal differentiation. Finally, we show that the ectopic expression of BLIMP1α in EBV-transformed cells can induce the viral lytic cycle. Our results suggest that LMP-1 expression in progenitor germinal center B cells could contribute to the pathogenesis of EBV-associated lymphomas by down-regulating BLIMP1α, in turn preventing plasma cell differentiation and induction of the viral lytic cycle.


2018 ◽  
Vol 9 ◽  
Author(s):  
Bárbara José Antunes Baptista ◽  
Alessandra Granato ◽  
Fábio B. Canto ◽  
Fabricio Montalvão ◽  
Lucas Tostes ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document