Abstract
Strong FOXP1 protein expression is a poor risk factor in diffuse large B-cell lymphoma and has been linked to an activated B-cell-like subtype, which preferentially expresses short FOXP1 (FOXP1S) proteins. However, both short isoform generation and function are incompletely understood. Here we prove by mass spectrometry and N-terminal antibody staining that FOXP1S proteins in activated B-cell-like diffuse large B-cell lymphoma are N-terminally truncated. Furthermore, a rare strongly FOXP1-expressing population of normal germinal center B cells lacking the N-terminus of the regular long protein (FOXP1L) was identified. Exon-targeted silencing and transcript analyses identified three alternate 5′ non-coding exons [FOXP1-Ex6b(s), FOXP1-Ex7b and FOXP1-Ex7c], downstream of at least two predicted promoters, giving rise to FOXP1S proteins. These were differentially controlled by B-cell activation and methylation, conserved in murine lymphoma cells, and significantly correlated with FOXP1S protein expression in primary diffuse large B-cell lymphoma samples. Alternatively spliced isoforms lacking exon 9 (e.g. isoform 3) did not encode FOXP1S, and an alternate long human FOXP1 protein (FOXP1AL) likely generated from a FOXP1-Ex6b(L) transcript was detected. The ratio of FOXP1L:FOXP1S isoforms correlated with differential expression of plasmacytic differentiation markers in U-2932 subpopulations, and altering this ratio was sufficient to modulate CD19 expression in diffuse large B-cell lymphoma cell lines. Thus, the activity of multiple alternate FOXP1 promoters to produce multiple protein isoforms is likely to regulate B-cell maturation.Introduction
Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease entity originating from germinal center (GC) or post-GC B cells such as plasmablasts.41 The majority of DLBCL can be classified according to cell-of-origin gene expression profile, as either germinal center (GC-DLBCL) or activated B-cell (ABC-DLBCL) subtype.95 While addition of rituximab to CHOP chemotherapy has improved DLBCL patients’ survival significantly,10 new therapies are needed for non-responding or relapsed patients (reviewed by Sehn and Gascoyne).11
Novel molecularly-targeted therapies are being sought particularly for the poorer prognosis ABC-DLBCL subtype following identification of key biological pathways contributing to disease pathogenesis, such as NF-κB pathway mutations and activation,1512 B-cell receptor (BCR) signaling,16 MALT1 activity,17 and BLIMP1 mutations.18 Maintenance of BCR signaling and prevention of plasma cell maturation to disrupt normal maturation/differentiation pathways is a common paradigm.
High FOXP1 expression correlates with the ABC-DLBCL subtype4 and poor clinical outcome in both the pre- and post-rituximab eras.2219 FOXP1 amplification and trisomy have been described in ABC-DLBCL,23 and translocations involving the immunoglobulin heavy chain (IGH) locus24 drive expression of a long ~75kDa FOXP1 protein (FOXP1L) that may contribute to GC-DLBCL tumor growth by potentiating Wnt/β-catenin signaling.25 Also, we have described abundant expression of short ~65kDa activation-induced FOXP1 proteins (FOXP1S) in ABC-DLBCL.26 Oncogenic activity of N-terminally truncated FOXP1 has been proposed following its truncation by an oncogenic virus27 and non-IGH translocations targeting the FOXP1 coding region in lymphoma.292824
Studies manipulating Foxp1 expression have established biological roles in early B-cell development3130 and in mature B cells.32 Direct FOXP1 target genes, including PAX5, PRDM1, and POU2F1, support a functional role in the GC reaction.32 Recent studies have provided insight into FOXP1 function in DLBCL cells, indicating contributions to proliferation,33 inhibition of apoptosis to compliment NF-κB-dependent regulation of proliferation,34 and Wnt signaling.25 However, further understanding of FOXP1 isoform expression and functionality is required to pursue emerging evidence indicating that FOXP1L and FOXP1S proteins have distinct functions.3122
Here we demonstrate that FOXP1S proteins in DLBCL are encoded by transcripts with alternate 5′ non-coding exons, not internal exon skipping.26 We identify a novel alternate long FOXP1 protein (designated FOXP1AL) and a GC B-cell population lacking the FOXP1L N-terminus. Isoform-biased FOXP1 depletion altering FOXP1L:FOXP1S stoichiometry regulates expression of CD19 in DLBCL cell lines, providing further evidence for isoforms having distinct functional roles in B-cell biology.
Methods
Cell culture
Diffuse large B-cell lymphoma and myeloma lines were sourced and cultured as described previously.26 COS-1, 293T and NIH-3T3 (ATCC) were cultured in DMEM containing 10% fetal bovine serum (Life Technologies, Paisley, UK), and murine B-cell lines 5TGM1-GFP (a gift of Claire Edwards, Oxford, UK) and A20 (ATCC) in RPMI containing 10% serum, 100 μM non-essential amino acids, 1 mM sodium pyruvate, 50 μM 2-mercaptoethanol.
Primary human samples
Diffuse large B-cell lymphoma patient samples were collected with informed consent in accordance with the Declaration of Helsinki, and the study was performed under local ethics committee approval from the Leeds West Regional Ethics Committee, Leeds, UK. RNA was isolated from DLBCL samples obtained prior to therapy as described.26 Primary B cells from blood buffy-coat preparations (National Blood Service, Bristol, UK) were activated with 50 μg/mL anti-IgM or 1:20,000 SAC plus 5 ng/mL recombinant human IL-2 (Sigma, St Louis, USA). Reactive tonsils were obtained with informed consent from John Radcliffe Hospital, Oxford, UK, and studies were conducted under ethical approval from NRES Committee South Central – Oxford B (C02.162).
Gene expression analyses
Random-primed cDNA was prepared from total RNA according to the manufacturer’s protocols (Life Technologies), and real-time PCR analyses performed by Chromo4 (BioRad, Herts., UK) using Express qPCR supermix or SYBR GreenER supermix (Life Technologies), and primers/probes as detailed in Online Supplementary Table S1. Standard RT-PCR to discriminate FOXP1-6b(L/S) transcripts used forward Ex6b(L)#1, Ex6b(L)#2, Ex6b(S), or control forward primers Ex6 or Ex8, all paired with reverse primer Ex10 (Online Supplementary Table S1). RNAseq analysis was performed on NCBI Gene Expression Omnibus (GEO) lymphoma dataset GSE50721.35
Immunoblot and immunoprecipitation
Whole cell extracts were prepared in RIPA, or nuclear and cytoplasmic extracts by commercial reagent (Affymetrix, High Wycombe, UK). Immunoprecipitations from 50 μg nuclear lysate were performed in IP buffer (1% Triton, 150 mM NaCl, 10 mM Tris, 1 mM EDTA) and complexes purified using μMACS Protein G microbeads (Miltenyi Biotec, Surrey, UK). Primary antibody details are provided in Online Supplementary Table S1.
Mass spectrometry
JC12 immunoprecipitates were subjected to parallel SDS-PAGE analyses for immunoblotting and silver staining. Stained bands corresponding to ~65kDa and ~75kDa FOXP1 proteins were excised, subjected to reduction, alkylation and trypsin digestion, and resulting peptides identified by mass spectrometry as previously described36 with formic acid modified to 0.1% in the gradient.
Immunohistochemistry
After formalin fixation, paraffin embedding and sectioning, slides were dewaxed and antigen retrieved by microwaving in 50 mM Tris, 2 mM EDTA pH9.0. Immunostaining with primary antibodies (Online Supplementary Table S1) was followed by secondary antibody and detection (Envision-Dako, Ely, UK).
Flow cytometry
Cells were labeled in PBS containing 0.5% bovine serum albumin, 2 mM EDTA with primary antibodies (Online Supplementary Table S1) and/or isotype controls, and/or secondary antibody (streptavidin APC, eBioscience) and data obtained by FACSCalibur (Becton Dickinson, San Jose, USA).
Transfection
For overexpression, cells were harvested 48 h after transfection with pcDNA4-HisMax expression vectors encoding human FOXP1-4 proteins using Lipofectamine (Life Technologies). For knockdown, cells were electroporated in the presence of 1 μM Stealth siRNA duplexes (Life Technologies) (Online Supplementary Table S1) using Amaxa Nucleofector (Lonza, Slough, UK) generally using Solution L, program X-001.
Results
Short FOXP1 proteins (FOXP1S) in ABC-DLBCL lack the N-terminus
We used mass spectrometry to characterize FOXP1S and FOXP1L proteins immunoprecipitated from GC- and ABC-DLBCL cell lines (Figure 1A and B). Peptides translated from most exons encoding the FOXP1L protein were identified, while peptides from the N-terminal coding exons (Ex), Ex6 and Ex7, were absent from FOXP1S proteins in ABC-DLBCL cell lines RIVA and OCI-Ly3 (Figure 1B). This is consistent with FOXP1S proteins deriving from transcripts where translation initiates in Ex8 (e.g. isoform 9)26 but inconsistent with internal deletion of Ex8 and/or Ex9 and/or Ex10 identified in FOXP1 isoforms 3, 5 and 8, which retain Ex6 and Ex7.26
To confirm N-terminal truncation of FOXP1 proteins in DLBCL, we validated a commercially available FOXP1 polyclonal antibody against an Ex7-encoded peptide. This antibody detected recombinant FOXP1L protein (Ex6-Ex21), but not FOXP1S protein (Ex8-Ex21), using both immunohistochemistry and Western blotting. However, there was cross-reactivity with both the related FOXP2 protein and an unknown, FOXP1 siRNA resistant, ~70kDa cytoplasmic protein in DLBCL cell lines (Online Supplementary Figure S1A–D). Thus, this reagent was used to study only nuclear expression in cell lines and tissues with known FOXP2 status. In DLBCL nuclear extracts, this N-terminal antibody recognized only FOXP1L, while our anti-C-terminal JC12 antibody detected both FOXP1S and FOXP1L proteins (Figure 1C). Thus FOXP1S proteins in multiple ABC-DLBCL cell lines lack the N-terminal epitopes encoded by Ex7.26 The N-terminal antibody was ineffective at routinely distinguishing ABC- versus GCB-DLBCL cell lines by immunohistochemistry (Online Supplementary Figure S2A), reflecting FOXP1L co-expression, non-specific cytoplasmic staining, and FOXP2 expression in RIVA. Interestingly, immuno-precipitation of FOXP1L using the N-terminal antibody co-immunoprecipitated FOXP1S, providing evidence for a physical FOXP1L-FOXP1S interaction in ABC-DLBCL (Online Supplementary Figure S1E).
A rare subset of germinal center B cells lack the FOXP1L N-terminus
The ABC-DLBCL cell-of-origin is proposed to be a plasmablastic B cell poised to exit the GC.41 Tonsillar B-cell follicles (FOXP2-negative) consistently exhibited comparable intensity and pattern of mantle zone staining when immunolabeled with N- and C-terminal FOXP1 antibodies (Figure 1D). However, rare GCs in some tonsils contained a small subpopulation of strongly FOXP1CD20 B cells (Figure 1D; inset) that were not effectively labeled using the N-terminal antibody. Thus, a small population of GC B cells may share the abundant FOXP1S protein expression observed in ABC-DLBCL. However, we cannot exclude an alternate FOXP1L protein (FOXP1AL), which we have identified in ABC-DLBCL.
Transcripts encoding FOXP1S proteins in ABC-DLBCL have variable 5′ non-coding exons and share coding exons 3′ from Ex8
N-terminally truncated FOXP1S proteins in ABC-DLBCL might derive from post-translational cleavage of the normal FOXP1L protein or alternate promoter usage. To distinguish these possibilities, we performed exon-targeted siRNA across the FOXP1 locus (Figure 2A), thus identifying transcripts producing FOXP1 proteins in ABC-DLBCL cell lines (RIVA and OCI-Ly3, and as a control the GC-DLBCL cell line DB) (Figure 2). FOXP1 coding exon targeting generally reduced FOXP1L levels, although this was sometimes difficult to detect in OCI-Ly3 due to low FOXP1L expression (Figure 2B). Consistent with siRNA targeting of the 5′ coding region being inefficient for some genes, Ex1–2 siRNA did not work at all, and Ex5 and Ex6 siRNAs targeted poorly. In contrast, targeting of FOXP1 Ex8 onwards silenced FOXP1 protein expression effectively, confirming coding function of the 3′ exons and the absence of FOXP1S coding transcripts with internal Ex8–9 deletions. Ex5 and Ex6 targeting had no effect on FOXP1S expression, suggesting that FOXP1S proteins were not post-translationally processed from FOXP1L.
Interestingly, two independent siRNAs targeting Ex7 that effectively silenced FOXP1L also partially depleted FOXP1S in both ABC-DLBCL cell lines (Figure 2B and C). As no Ex7-encoded peptides were identified in FOXP1S by mass spectrometry, a proportion of FOXP1S-coding transcripts may contain a non-coding Ex7. Indeed one such transcript, with transcription starting in the 3′ end of alternative Ex6b(S) is described (Figure 3). Thus FOXP1S-coding transcripts in ABC-DLBCL share common 3′ exons (from exon 8 onwards), have variable 5′ non-coding exons, and are not encoded by previously reported splice variants26 lacking exons 8, 9 and/or 10.
DLBCL cell lines expressing FOXP1S protein transcribe multiple 5′ alternate exon-containing FOXP1 mRNA species
To explore the relationship between FOXP1 proteins and transcripts, panels of GC- and ABC-DLBCL lines were ranked by increasing FOXP1S:FOXP1L protein expression ratio (Online Supplementary Figure S2B). Based on our data, additional database searching, and published FOXP1 RNA-sequencing data from primary DLBCL biopsies,24 we assembled a list of FOXP1 transcripts with the potential to encode FOXP1L and FOXP1S proteins in ABC-DLBCL (Figure 3A). There appear to be two transcriptional start sites within Ex6b, with the 5′ longer Ex6b transcript (Ex6b(L)) predicted to encode a long FOXP1 protein with an alternate N-terminus (FOXP1AL), while the shorter (Ex6b(S)) initiates translation from Ex8, as do transcripts containing other alternate 5′ non-coding exons Ex7b and Ex7c.
Real-time PCR analysis of common 3′ exons (Ex20–21) demonstrated increased FOXP1 expression in most ABC-DLBCL cell lines, while expression of 5′ Ex6–7 was variable (Figure 3B). Alternate exons Ex7b and Ex7c were preferentially transcribed in ABC-DLBCL cell lines, while Ex6b (encoding FOXP1L or FOXP1S proteins) was only slightly more abundant in ABC-DLBCL lines (Figure 3C). RT-PCR analysis demonstrated that, in contrast to Ex6b(S), Ex6b(L) was abundant only in the ABC-DLBCL cell line HBL-1 (Online Supplementary Figure S3A). Interestingly, reduced recognition of long FOXP1 by the N-terminal antibody in HBL-1 (Figure 1C) is consistent with expression of an additional long FOXP1 protein containing an alternate N-terminus, FOXP1AL. Transfection confirmed that Ex6b(L) encodes only the FOXP1AL protein (Online Supplementary Figure S1F).
Increased expression of alternate 5′ Ex6b and Ex7b transcripts in ABC-DLBCL versus GC-DLBCL lines was also detectable by RNA-sequencing (Figure 4 and Online Supplementary Table S2), although only the Ex6b(L) increase was significant. Ex7c was not detected by this method, reflecting its relatively low abundance. In contrast to previously reported RT-PCR data,26 Ex4a was not reliably increased in ABC-DLBCL (Figure 4 and Online Supplementary Figure S3B). Thus, no single alternate transcript was specifically associated with increased FOXP1S expression, and almost all ABC-DLBCL cell lines expressed multiple alternate transcripts.
FOXP1 transcripts with alternate exons 6 and 7 correlate with FOXP1S protein expression in primary human DLBCL
The expression of FOXP1 total transcripts and those with alternate 5′ exons was further validated in a panel of primary DLBCL cases with known FOXP1L and FOXP1S protein expression, determined previously by immunoblotting.26 Expression of FOXP1 transcripts containing Ex6b or Ex7c was significantly increased in tumors where FOXP1S protein levels were equivalent to or exceeded FOXP1L, while there was no relationship with total FOXP1 transcripts (Figure 5A). Expression of Ex6b, Ex7b, Ex7c and total FOXP1 was significantly higher in non-GC-DLBCL (Figure 5B). Combining expression profiles of all three FOXP1S-encoding transcripts did not improve the accuracy of predicting GC or non-GC status (data not shown); some non-GC-DLBCL exhibited low expression of them all. Expression of Ex7b and Ex7c transcripts was significantly related to the intensity of tumoral nuclear FOXP1 protein positivity determined by immunohistochemistry (Figure 5C). Thus, despite lower relative abundance in DLBCL cell lines, only Ex7c-containing transcript levels correlate significantly with both FOXP1 protein expression levels, DLBCL cell-of-origin subtype, and predominant FOXP1S protein expression in primary tumors. Ex4a expression was not significantly related to FOXP1 protein or GC/non-GC status in primary DLBCL (Online Supplementary Figure S3C).
Malignant mature murine B cells express Foxp1S protein and N-terminally truncated Foxp1 transcripts partially conserved with mature human B cells
Cross-species conservation supports biological significance, and a murine Foxp1S isoform termed Foxp1D (initiating exon originally designated mEx2b,37 revised to mEx5b)38 has been described.37 Multiple transcripts with alternate non-coding 5′ exons encoding murine Foxp1S protein in a murine B-cell lymphoma model, A20, that expresses both Foxp1L and Foxp1S proteins were investigated (Online Supplementary Figure S4A). Comparisons of the murine Foxp1 genomic sequence with alternate 5′ exons in human FOXP1 transcripts identified potential murine exons with homology to human Ex6b (mEx4b), Ex7b (mEx5b) and Ex7c (mEx5c) (Online Supplementary Figure S4B and C). However, the revised mEx2b alternate 5′ exon (predicted to encode Foxp1L or Foxp1A)37 had only low similarity to human Ex4a sequences. Importantly, expression of Foxp1S-coding murine transcripts containing alternate 5′ exon mEx4b or mEx5b was detectable in the malignant mature B-cell lines A20 and 5TGM1 and associated with reduced expression of Foxp1L Ex2–4 transcripts (Online Supplementary Figure S4D). Despite the significance of Ex7c in human DLBCL, transcripts containing its potential murine equivalent (mEx5c) could not be reliably detected. In summary, multiple transcripts with alternate 5′ exons contribute to FOXP1S protein expression in both murine and human lymphoma.
FOXP1 alternate 5′ exon usage is differentially induced by activation/maturation stimuli
The redundancy of multiple alternate 5′ exons giving rise to FOXP1S may enable several pathways (potentially different cell types or developmental stages) to fine-tune FOXP1S protein expression. Supporting this hypothesis, activation of primary human naïve B cells from 2 individuals with multiple stimuli, previously shown to induce FOXP1S protein,26 increased expression of Ex6b(L) Ex7b and Ex7c, but had little effect on total Ex6b transcripts (Figure 6A). The latter indicates relatively high expression of Ex6b(S) versus Ex6b(L), as in DLBCL cell lines (Online Supplementary Figure S3A). Activation of B-cell-derived cell lines induced Ex7c but not total Ex6b or reliably Ex7b expression, while Ex6b transcripts were induced only in the myeloid cell line HL-60, despite successful activation of other myeloid lines (Figure 6B and Online Supplementary Figure S5A and B). Lack of Ex6b(L) transcripts (Figure 6B; inset) indicates this Ex6b to be FOXP1S-coding. Thus, multiple conserved FOXP1 transcripts with alternate 5′ exons are induced by activation/maturation events in a cell-type specific manner to produce FOXP1S protein. Treatment of several GC-DLBCL cell lines with 5-azacytidine increased Ex7b and Ex7c but not Ex6b expression (Online Supplementary Figure S5C), implicating methylation as another mechanism differentially regulating alternate Foxp1 promoter activity between DLBCL subtypes.
FOXP1S:FOXP1L ratio controls CD19 expression in human DLBCL
As GC- and ABC-DLBCL differ in their putative cell-of-origin, the relationship between FOXP1S:FOXP1L ratio and expression of B-cell activation/maturation cell surface markers was investigated further across DLBCL cell lines. Expression of the B-cell marker CD20 was uniformly robust, and that of the plasma cell marker CD138 uniformly weak or absent (Figure 7A). In contrast, surface expression of CD19, CD45 (B220 isoform), and CD27 generally decreased as the FOXP1S to FOXP1L expression ratio increased (Figure 7A), with relatively few exceptions (e.g. CD19 in DB and CD45 in Karpas-422). Cell lines such as SU-DHL-4 and U-2932 exhibited both intermediate FOXP1S:FOXP1L ratios and intermediate expression of the immaturity/memory markers CD19, B220 and/or CD27. Importantly, in these ‘intermediate’ DLBCL cell lines, transfection of siRNAs targeting Ex7 to increase the FOXP1S:FOXP1L ratio (Figure 2), but not targeting of total FOXP1, significantly reduced CD19 expression (Figure 7B and C).
The ABC-DLBCL cell line U-2932 has two subpopulations ‘R1’ and ‘R2’ (Figure 8A) both present as ‘clones’ in the original patient (CD20CD38 and CD20CD38, respectively), which can be maintained stably and display both common and unique genetic aberrations.39 Purified R1 and R2 populations exhibited clumped versus single-cellular growth habits, respectively (Figure 8B), and R1 showed higher FOXP1S protein expression and increased expression of FOXP1 Ex6b, Ex7b and Ex7c transcripts (Figure 8C and D). Furthermore, R1 has elevated expression of plasmablastic markers (IRF4, XBP1) and reduced expression of the FOXP1S-repressed target gene HIP1R22 (Figure 8D), again demonstrating a positive association between FOXP1S and B-cell maturity.
Discussion
Characterization of FOXP1 isoform complexity in both human and murine lymphoma cells is important in order to understand their roles in lymphoma biology and clinical relevance.
We have proven our original hypothesis26 that FOXP1S proteins in ABC-DLBCL lack the N-terminus using both mass spectrometry and a commercially available polyclonal N-terminal antibody. An improved N-terminal antibody, lacking the non-FOXP1 cytoplasmic cross-reactivity and nuclear FOXP2 cross-reactivity seen with the current antibody, might routinely identify strongly JC12 (FOXP1 C-terminal epitope) DLBCL that lack the N-terminus. Despite its limitations, the current polyclonal antibody may be of interest within the research community, e.g. to validate expression of C-terminally truncated FOXP1 proteins, such as the FOXP1w isoform up-regulated by SF3B1 mutation in poor prognosis B-cell chronic lymphocytic leukemia.
Western blotting data have demonstrated that normal lymphoid tissue and the microenvironment surrounding FOXP1-negative tumors express predominantly FOXP1L protein(s).26 Thus, our identification of a rare GC B-cell population with high-level expression of FOXP1 protein(s) lacking Ex7-encoded epitopes is particularly interesting. Additional reagents (e.g. antibodies to novel epitopes in the N-terminus of FOXP1AL) will help to definitively characterize this strongly JC12 population and examine whether FOXP1AL or FOXP1S isoforms predominate. Such studies may help to define a normal counterpart for ABC-DLBCL. FOXP1 exon-targeted siRNA and expression studies have identified at least three distinct FOXP1 proteins in DLBCL; FOXP1L, FOXP1AL (long forms) and FOXP1S, the latter two being primarily expressed in ABC-DLBCL. FOXP1AL detection in the HBL-1 ABC-DLBCL cell line is consistent with published transcript data describing potential expression of this isoform (FOXP1-011, ENST00000491238) in primary DLBCL.2624 Since all alternate 5′ exons were targeted inefficiently by siRNA in DLBCL cells, methods such as FOXP1 locus editing could help to determine particular transcript contributions. Importantly, transcript conservation in murine lymphoma cells (e.g. human Ex6b and murine equivalent mEx4b being most abundant) should simplify Foxp1S functional studies. While no individual 5′ non-coding exon defines FOXP1S protein expression in primary DLBCL, Ex7c remains the best single transcript predictor and could be a useful addition to prognostic/diagnostic gene panels for DLBCL. Our data suggest that the FOXP1S/L ratio may also help define the stage of developmental block in DLBCL and improve stratification of DLBCL subgroups.
Although FOXP1 proteins expressed in malignant and normal B cells are similar, the control of FOXP1 expression appears distinct. Genetic abnormalities truncating FOXP1 are infrequent,292824 and our data indicate the majority of N-terminally-deleted FOXP1 expression in lymphoma is generated by alternate internal 5′ promoter usage (Ex6b being particularly abundant). Thus, it will be important to understand the likely oncogenic mechanisms controlling these transcriptional events. Significantly, BLIMP1 has been shown to bind the FOXP1 locus,40 and thus BLIMP1 mutation18 may drive ABC-DLBCL lymphomagenesis at least in part by mis-regulating FOXP1 isoform expression.
Historically, many clinical studies have not evaluated heterogeneity of marker expression across areas of a tumor. However, studies of clonal evolution have highlighted the importance of heterogeneity in a variety of malignancies, particularly in developing treatment resistance. We previously noted in a lymphoma biopsy with FOXP1 translocation that the localized area of tumor containing the translocation had stronger FOXP1 protein expression ((PJ Brown et al, unpublished data, 2014). Here we demonstrate that subpopulations from a single DLBCL patient (present in U-2932) exhibit distinct FOXP1 isoform patterns alongside different immunophenotypes and expression of transcription factors involved in B-cell differentiation. Greater understanding of FOXP1 isoform heterogeneity may help to identify therapy-resistant DLBCL clones, as elevated FOXP1 is linked to resistance in response to both CHOP-R and rituximab monotherapy in lymphoma.41222119
Abundant Ex6bS transcript expression in GC-DLBCL cell lines lacking FOXP1S protein expression suggests post-transcriptional regulatory mechanisms may also regulate FOXP1S protein expression in DLBCL. Alternate exon-specific miRNA activity, in addition to the previously reported global miRNA targeting of FOXP1,3331 could be one regulatory mechanism, but there are many possibilities. Our finding that the first methionine codon in hEx6b(L) enabling translation of FOXP1AL is not conserved in mouse must be considered in studies of murine lymphoma, and may also prove significant outside of lymphoma and B-cell biology. Particularly the established neuronal roles of FOXP1,42 emerging evidence for FOXP1 alterations in human behavioral disorders,43 and the proposed contribution of related FOXP2 sequence evolution to language acquisition44 indicate that neuronal studies of human-specific FOXP1AL may be warranted.
Total FOXP1 depletion studies indicate a contribution to cell viability and regulation of developmental target genes such as PAX5 and PRDM1 in lymphoma cells.45333225 In addition, Dekker et al. have recently shown that total FOXP1 activates nearly all BCR-dependent genes; however, as FOXP1 silencing did not decrease BCR clustering, they concluded that it contributed to chronic B-cell receptor signaling (CABS) but only minimally to CABS-directed NF-κB activation.46 Our findings, showing no decrease in CD19 expression upon total FOXP1 silencing, are consistent with these data. Interestingly, an siRNA that preferentially silenced FOXP1L was recently reported to be less effective at inducing cell death than total FOXP1 targeting in multiple DLBCL cell lines.45 While the same study generally observed stronger regulation of gene expression on silencing all FOXP1 isoforms, there were some exceptions.47 Our isoform-biased depletion studies presented here indicate that, in accordance with our previous findings (where the FOXP1 target gene HIP1R was preferentially regulated in ABC-DLBCL),22 and complimentary to some common functions,47 FOXP1L and FOXP1S have at least some distinct functional properties, including CD19 regulation. CD19 is a critical regulator of BCR signaling48 via both BCR-dependent and independent mechanisms, thus FOXP1 isoforms (rather than total expression levels) may contribute to surface BCR activity. Distinct long and short FOXP1 functions are highly likely to derive from altered protein-protein interactions at the N-terminus. Given that induction of FOXP1S expression, for example during B-cell activation, is associated with many other changes, the cellular environment is likely to play a crucial role in defining long and short FOXP1 functions.
Overall, we favor a model in which temporally- or spatially-distinct FOXP1L and FOXP1S expression have some conserved functions but mediate also distinct isoform-specific functions during mature B-cell development and in lymphoma pathogenesis. CD19 regulation may provide a potential mechanism for the reported FOXP1 regulation of BCR signaling,4946 and modulating FOXP1 could, therefore, have clinical benefit for therapies targeting CD19 in DLBCL, e.g. anti-CD19 chimeric antigen receptors.50 Studying expression, regulation and function of FOXP1 isoforms in relation to developmental blocks in DLBCL may also lead to the identification of novel therapeutic differentiation strategies for this disease.
Acknowledgments
The authors would like to acknowledge the important contributions from Dr Philip Brown, who sadly died before the completion of this study and drafting of the manuscript. We thank Dr Helen Ferry for cell sorting and Aengus Stewart at The Francis Crick Institute for analysis of HiSeq data.
Footnotes
- ↵* PJB and DMG contributed equally to this work.
- ↵† Deceased, 2nd March 2014
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/101/7/861
- FundingPJB, DMG, LL, SLF and HS were supported by Specialist Program Grants (Ref:10044, 13047) to AHB from Bloodwise and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre program. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health. NM and DPC were supported by core funding from The Francis Crick Institute, and a MRC career development award MR/J008060/1 to DPC.
- Received January 15, 2016.
- Accepted April 4, 2016.
References
- Kuppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer. 2005; 5(4):251-262. PubMedhttps://doi.org/10.1038/nrc1589Google Scholar
- Lenz G, Staudt LM. Aggressive lymphomas. N Engl J Med. 2010; 362(15):1417-1429. PubMedhttps://doi.org/10.1056/NEJMra0807082Google Scholar
- Pasqualucci L, Dalla-Favera R. SnapShot: diffuse large B cell lymphoma. Cancer Cell. 2014; 25(1):132-132. PubMedhttps://doi.org/10.1016/j.ccr.2013.12.012Google Scholar
- Shaffer AL, Rosenwald A, Staudt LM. Lymphoid malignancies: the dark side of B-cell differentiation. Nat Rev Immunol. 2002; 2(12):920-932. PubMedhttps://doi.org/10.1038/nri953Google Scholar
- Alizadeh AA, Eisen MB, Davis RE. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000; 403(6769):503-511. PubMedhttps://doi.org/10.1038/35000501Google Scholar
- Care MA, Barrans S, Worrillow L. A microarray platform-independent classification tool for cell of origin class allows comparative analysis of gene expression in diffuse large B-cell lymphoma. PLoS One. 2013; 8(2):e55895. PubMedhttps://doi.org/10.1371/journal.pone.0055895Google Scholar
- Lossos IS, Morgensztern D. Prognostic biomarkers in diffuse large B-cell lymphoma. J Clin Oncol. 2006; 24(6):995-1007. PubMedhttps://doi.org/10.1200/JCO.2005.02.4786Google Scholar
- Rosenwald A, Wright G, Chan WC. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med. 2002; 346(25):1937-1947. PubMedhttps://doi.org/10.1056/NEJMoa012914Google Scholar
- Wright G, Tan B, Rosenwald A. A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc Natl Acad Sci USA. 2003; 100(17):9991-9996. PubMedhttps://doi.org/10.1073/pnas.1732008100Google Scholar
- Dupire S, Coiffier B. Targeted treatment and new agents in diffuse large B cell lymphoma. Int J Hematol. 2010; 92(1):12-24. PubMedhttps://doi.org/10.1007/s12185-010-0609-6Google Scholar
- Sehn LH, Gascoyne RD. Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity. Blood. 2015; 125(1):22-32. PubMedhttps://doi.org/10.1182/blood-2014-05-577189Google Scholar
- Compagno M, Lim WK, Grunn A. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009; 459(7247):717-721. PubMedhttps://doi.org/10.1038/nature07968Google Scholar
- Dunleavy K, Pittaluga S, Czuczman MS. Differential efficacy of bortezomib plus chemotherapy within molecular subtypes of diffuse large B-cell lymphoma. Blood. 2009; 113(24):6069-6076. PubMedhttps://doi.org/10.1182/blood-2009-01-199679Google Scholar
- Lenz G, Davis RE, Ngo VN. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008; 319(5870):1676-1679. PubMedhttps://doi.org/10.1126/science.1153629Google Scholar
- Ngo VN, Davis RE, Lamy L. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006; 441(7089):106-110. PubMedhttps://doi.org/10.1038/nature04687Google Scholar
- Davis RE, Ngo VN, Lenz G. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010; 463(7277):88-92. PubMedhttps://doi.org/10.1038/nature08638Google Scholar
- Ferch U, Kloo B, Gewies A. Inhibition of MALT1 protease activity is selectively toxic for activated B cell-like diffuse large B cell lymphoma cells. J Exp Med. 2009; 206(11):2313-2320. PubMedhttps://doi.org/10.1084/jem.20091167Google Scholar
- Mandelbaum J, Bhagat G, Tang H. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma. Cancer Cell. 2010; 18(6):568-579. PubMedhttps://doi.org/10.1016/j.ccr.2010.10.030Google Scholar
- Banham AH, Connors JM, Brown PJ. Expression of the FOXP1 transcription factor is strongly associated with inferior survival in patients with diffuse large B-cell lymphoma. Clin Cancer Res. 2005; 11(3):1065-1072. PubMedGoogle Scholar
- Barrans SL, Fenton JA, Ventura R. Deregulated over expression of FOXP1 protein in diffuse large B-cell lymphoma does not occur as a result of gene rearrangement. Haematologica. 2007; 92(6):863-864. PubMedhttps://doi.org/10.3324/haematol.10656Google Scholar
- Nyman H, Jerkeman M, Karjalainen-Lindsberg ML, Banham AH, Leppa S. Prognostic impact of activated B-cell focused classification in diffuse large B-cell lymphoma patients treated with R-CHOP. Mod Pathol. 2009; 22(8):1094-1101. PubMedhttps://doi.org/10.1038/modpathol.2009.73Google Scholar
- Wong KK, Gascoyne DM, Brown PJ. Reciprocal expression of the endocytic protein HIP1R and its repressor FOXP1 predicts outcome in R-CHOP-treated diffuse large B-cell lymphoma patients. Leukemia. 2014; 28(2):362-372. PubMedhttps://doi.org/10.1038/leu.2013.224Google Scholar
- Lenz G, Wright GW, Emre NC. Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways. Proc Natl Acad Sci USA. 2008; 105(36):13520-13525. PubMedhttps://doi.org/10.1073/pnas.0804295105Google Scholar
- Rouhigharabaei L, Finalet Ferreiro J, Tousseyn T. Non-IG aberrations of FOXP1 in B-cell malignancies lead to an aberrant expression of N-truncated isoforms of FOXP1. PLoS One. 2014; 9(1):e85851. PubMedhttps://doi.org/10.1371/journal.pone.0085851Google Scholar
- Walker MP, Stopford CM, Cederlund M. FOXP1 potentiates Wnt/beta-catenin signaling in diffuse large B cell lymphoma. Sci Signal. 2015; 8(362):ra12. PubMedhttps://doi.org/10.1126/scisignal.2005654Google Scholar
- Brown PJ, Ashe SL, Leich E. Potentially oncogenic B-cell activation-induced smaller isoforms of FOXP1 are highly expressed in the activated B cell-like subtype of DLBCL. Blood. 2008; 111(5):2816-2824. PubMedhttps://doi.org/10.1182/blood-2007-09-115113Google Scholar
- Pajer P, Pecenka V, Kralova J. Identification of potential human oncogenes by mapping the common viral integration sites in avian nephroblastoma. Cancer Res. 2006; 66(1):78-86. PubMedhttps://doi.org/10.1158/0008-5472.CAN-05-1728Google Scholar
- Goatly A, Bacon CM, Nakamura S. FOXP1 abnormalities in lymphoma: translocation breakpoint mapping reveals insights into deregulated transcriptional control. Mod Pathol. 2008; 21(7):902-911. PubMedhttps://doi.org/10.1038/modpathol.2008.74Google Scholar
- Wlodarska I, Veyt E, De Paepe P. FOXP1, a gene highly expressed in a subset of diffuse large B-cell lymphoma, is recurrently targeted by genomic aberrations. Leukemia. 2005; 19(8):1299-1305. PubMedhttps://doi.org/10.1038/sj.leu.2403813Google Scholar
- Hu H, Wang B, Borde M. Foxp1 is an essential transcriptional regulator of B cell development. Nat Immunol. 2006; 7(8):819-826. PubMedhttps://doi.org/10.1038/ni1358Google Scholar
- Rao DS, O’Connell RM, Chaudhuri AA. MicroRNA-34a perturbs B lymphocyte development by repressing the forkhead box transcription factor Foxp1. Immunity. 2010; 33(1):48-59. PubMedhttps://doi.org/10.1016/j.immuni.2010.06.013Google Scholar
- Sagardoy A, Martinez-Ferrandis JI, Roa S. Downregulation of FOXP1 is required during germinal center B-cell function. Blood. 2013; 121(21):4311-4320. PubMedhttps://doi.org/10.1182/blood-2012-10-462846Google Scholar
- Craig VJ, Cogliatti SB, Imig J. Myc-mediated repression of microRNA-34a promotes high-grade transformation of B-cell lymphoma by dysregulation of FoxP1. Blood. 2011; 117(23):6227-6236. PubMedhttps://doi.org/10.1182/blood-2010-10-312231Google Scholar
- van Keimpema M, Gruneberg LJ, Mokry M. FOXP1 directly represses transcription of proapoptotic genes and cooperates with NF-kappaB to promote survival of human B cells. Blood. 2014; 124(23):3431-3440. PubMedhttps://doi.org/10.1182/blood-2014-01-553412Google Scholar
- Hardee J, Ouyang Z, Zhang Y. STAT3 targets suggest mechanisms of aggressive tumorigenesis in diffuse large B-cell lymphoma. G3 (Bethesda). 2013; 3(12):2173-2185. PubMedhttps://doi.org/10.1534/g3.113.007674Google Scholar
- Hooper C, Sainz-Fuertes R, Lynham S. Wnt3a induces exosome secretion from primary cultured rat microglia. BMC Neurosci. 2012; 13:144. PubMedhttps://doi.org/10.1186/1471-2202-13-144Google Scholar
- Wang B, Lin D, Li C, Tucker P. Multiple domains define the expression and regulatory properties of Foxp1 forkhead transcriptional repressors. J Biol Chem. 2003; 278(27):24259-24268. PubMedhttps://doi.org/10.1074/jbc.M207174200Google Scholar
- Gabut M, Samavarchi-Tehrani P, Wang X. An alternative splicing switch regulates embryonic stem cell pluripotency and reprogramming. Cell. 2011; 147(1):132-146. PubMedhttps://doi.org/10.1016/j.cell.2011.08.023Google Scholar
- Quentmeier H, Amini RM, Berglund M. U-2932: two clones in one cell line, a tool for the study of clonal evolution. Leukemia. 2013; 27(5):1155-1164. PubMedhttps://doi.org/10.1038/leu.2012.358Google Scholar
- Doody GM, Care MA, Burgoyne NJ. An extended set of PRDM1/BLIMP1 target genes links binding motif type to dynamic repression. Nucleic Acids Res. 2010; 38(16):5336-5350. PubMedhttps://doi.org/10.1093/nar/gkq268Google Scholar
- Rubenstein JL, Fridlyand J, Abrey L. Phase I study of intraventricular administration of rituximab in patients with recurrent CNS and intraocular lymphoma. J Clin Oncol. 2007; 25(11):1350-1356. PubMedhttps://doi.org/10.1200/JCO.2006.09.7311Google Scholar
- Rousso DL, Gaber ZB, Wellik D, Morrisey EE, Novitch BG. Coordinated actions of the forkhead protein Foxp1 and Hox proteins in the columnar organization of spinal motor neurons. Neuron. 2008; 59(2):226-240. PubMedhttps://doi.org/10.1016/j.neuron.2008.06.025Google Scholar
- O’Roak BJ, Deriziotis P, Lee C. Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet. 2011; 43(6):585-589. PubMedhttps://doi.org/10.1038/ng.835Google Scholar
- Fisher SE, Ridley M. Evolution. Culture, genes, and the human revolution. Science. 2013; 340(6135):929-930. PubMedhttps://doi.org/10.1126/science.1236171Google Scholar
- Flori M, Schmid CA, Sumrall ET. The hematopoietic oncoprotein FOXP1 promotes tumor cell survival in diffuse large B-cell lymphoma by repressing S1PR2 signaling. Blood. 2016; 127(11):1438-1448. PubMedhttps://doi.org/10.1182/blood-2015-08-662635Google Scholar
- Dekker JD, Park D, Shaffer AL. Subtype-specific addiction of the activated B-cell subset of diffuse large B-cell lymphoma to FOXP1. Proc Natl Acad Sci USA. 2016; 113(5):E577-586. PubMedhttps://doi.org/10.1073/pnas.1524677113Google Scholar
- Wang H, Geng J, Wen X. The transcription factor Foxp1 is a critical negative regulator of the differentiation of follicular helper T cells. Nat Immunol. 2014; 15(7):667-675. PubMedhttps://doi.org/10.1038/ni.2890Google Scholar
- Depoil D, Fleire S, Treanor BL. CD19 is essential for B cell activation by promoting B cell receptor-antigen microcluster formation in response to membrane-bound ligand. Nat Immunol. 2008; 9(1):63-72. PubMedhttps://doi.org/10.1038/ni1547Google Scholar
- Mraz M, Chen L, Rassenti LZ. miR-150 influences B-cell receptor signaling in chronic lymphocytic leukemia by regulating expression of GAB1 and FOXP1. Blood. 2014; 124(1):84-95. PubMedhttps://doi.org/10.1182/blood-2013-09-527234Google Scholar
- Kochenderfer JN, Dudley ME, Kassim SH. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015; 33(6):540-549. PubMedhttps://doi.org/10.1200/JCO.2014.56.2025Google Scholar