Subtype-specific addiction of the activated B-cell subset of diffuse large B-cell lymphoma to FOXP1

JD Dekker, D Park, AL Shaffer III… - Proceedings of the …, 2016 - National Acad Sciences
Proceedings of the National Academy of Sciences, 2016National Acad Sciences
High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the
aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the
better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with
poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however,
remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL
cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional …
High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however, remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional enforcement of ABC-DLBCL hallmarks, including the classical NF-κB and MYD88 (myeloid differentiation primary response gene 88) pathways. FOXP1 promoted gene expression underlying transition of the GCB cell to the plasmablast—the transient B-cell stage targeted in ABC-DLBCL transformation—by antagonizing pathways distinctive of GCB-DLBCL, including that of the GCB “master regulator,” BCL6 (B-cell lymphoma 6). Cell-line derived FOXP1 target genes that were highly correlated with FOXP1 expression in primary DLBCL accurately segregated the corresponding clinical subtypes of a large cohort of primary DLBCL isolates and identified conserved pathways associated with ABC-DLBCL pathology.
National Acad Sciences