Transcriptome sequencing identifies novel fusion transcripts in PTCL-NOS
To identify fusion events in PTCL-NOS, we performed high coverage, paired-end RNA sequencing (RNA-seq) on a cohort of 15 PTCL-NOS (Supplementary Table 1) cases and 3 PTCL-TFH13. Gene expression analysis indicated expression of T follicular helper (Tfh) markers in the PTCL-TFH cases (Supplementary Fig. 1a). In agreement with the histopathological diagnosis, a subset of PTCL-NOS cases expressed TNFRSF8 (CD30). Expression of the ALCL marker genes BATF3 and TMOD115 clearly separated the PTCL-NOS cases from ALCL cases12 (Supplementary Fig. 1b).
In 5 cases, we detected fusion transcripts, including the VAV1-MYO1F and TBL1XR1-TP63 fusions that were reported previously in PTCL-NOS11,16, and further confirm the validity of this cohort. In addition, we identified 3 novel in-frame fusion transcripts: FYN-TRAF3IP2 (2 cases), KHDRBS1-LCK (1 case) and SIN3A-FOXO1 (1 case) (Supplementary Fig. 1c). We found evidence for a gene fusion between the neighboring FYN and TRAF3IP2 genes in 1/15 PTCL-NOS (case PTCL2) and 1/3 PTCL-TFH (case FTCL4). In the PTCL-NOS case, exon 7 of FYN was fused to exon 3 of TRAF3IP2 (Fig. 1a). To exclude that such fusion was the result of transcriptional read-through, we investigated the genomic region by long range PCR and we identified an interstitial deletion of 92597 bp on chromosome 6 (Fig. 1b). In the PTCL-TFH case, exon 8 of FYN was fused to exon 3 of TRAF3IP2 (Fig. 1a). In both variants of the fusion protein, the FYN moiety contained the membrane localization motif (SH4 domain) and the SH3 domain of FYN, but lacked the FYN tyrosine kinase domain. The TRAF3IP2 moiety consisted of the almost complete open reading frame, only lacking the first 7 amino acids and thus retaining the TNF receptor associated factor 6 (TRAF6) binding domain in the fusion protein. Additionally, we discovered a KHDRBS1-LCK gene fusion that was caused by an interstitial deletion on chromosome 1 in 1/15 PTCL-NOS (case PTCL17). This gene fusion generated a chimeric transcript in which exon 3 of KHDRBS1 was fused to exon 3 of LCK, resulting in the fusion of the QUA1 and K homology (KH) dimerization domains of KHDRBS117 to the SH3, SH2 and kinase domains of LCK (Fig. 1c).
To validate the importance of these fusion genes, we tested the presence of the FYN-TRAF3IP2 and the KHDRBS1-LCK fusion genes with RT-PCR in an independent validation cohort of 37 PTCL cases (30 PTCL-NOS, 6 PTCL-TFH, 1 Adult T-cell leukemia/lymphoma (ATLL)). This screen confirmed that the FYN-TRAF3IP2 fusion is highly recurrent as it was detected (and verified by Sanger sequencing) in 6/37 (3 PTCL-NOS, 3 PTCL-TFH) samples (Supplementary Fig. 1d-e, Supplementary Table 2). We detected no additional cases of the KHDRBS1-LCK fusion in the independent validation cohort.
FYN-TRAF3IP2-dependent signaling intersects with TCR signaling
To study the oncogenic properties of the FYN-TRAF3IP2 fusion protein, we cloned the open reading frame (ORF) derived from patient cDNA (case PTCL2) in a bicistronic pMSCV vector with an IRES-GFP reporter (pMIG). As controls, full length TRAF3IP2 and the N-terminal fragment of FYN containing the membrane localization motif, the SH3 domain and a truncated SH2 domain (FYN1–232) were cloned in the pMIG vector (Supplementary Fig. 2a). Only retroviral transduction of interleukin-3 (IL-3) dependent Ba/F3 cells with the FYN-TRAF3IP2 ORF conferred IL-3 independent growth in vitro, indicating that the FYN-TRAF3IP2 fusion protein is endowed with oncogenic properties and that the combined signaling properties of both fusion partners are required (Fig. 2a).
The oncogenic signaling properties of the FYN-TRAF3IP2 fusion protein could not be related to tyrosine kinase signaling of the FYN kinase domain since it was not retained in the fusion protein. Instead, it was only the N-terminal part of FYN that was fused to the majority of the TRAF3IP2 protein. Under physiological circumstances, TRAF3IP2 is essential for IL-17 signaling. Upon engagement of the heterodimeric IL-17RA/IL-17RC complex, TRAF3IP2 translocates to the juxtamembrane compartment via homotypic SEFIR domain interactions and relays signals to the canonical NF-κB pathway and mitogen-activated protein kinase (MAPK) pathways18. IL-17RA is ubiquitously expressed, but IL-17RC expression is more restricted and defines the cellular response to IL-1718. Neither T cells from healthy volunteers (Supplementary Fig. 2b), nor any of the lymphoma samples in our cohort (Supplementary Fig. 2c) expressed IL17RC. Accordingly, the levels of TRAF3IP2-regulated transcripts in naive CD4+ T cells transduced with empty pMIG vector (EV), pMIG-TRAF3IP2 or pMIG-FYN-TRAF3IP2 were primarily determined by expression of FYN-TRAF3IP2. Stimulation with IL-17 had an additive rather than a synergistic effect on the expression TRAF3IP2-regulated transcripts (Fig. 2b). Combined, these data strongly suggest that the biological activity of the FYN-TRAF3IP2 fusion protein did not depend on IL-17 signaling.
Given that TCR signaling is a nexus in the integrated control of proliferation and survival in healthy and malignant T cells and TRAF3IP2 activates NF-kB signaling and Mitogen-Activated Protein Kinase (MAPK) downstream of the IL-17 receptor, we hypothesized that aberrant FYN-TRAF3IP2 signaling would intersect with NF-kB signaling and MAPK signaling downstream of the TCR. We transduced the Jurkat T ALL cell line to induce the expression of TRAF3IP2 or FYN-TRAF3IP2. Western blot analysis revealed increased Ser536 phosphorylation of the NF-kB subunit p65 (also known as RelA), indicative of increased activation of the canonical NF-kB pathway. Processing of p100 to p52 and Ser866/870 phosphorylation of p100 did not provide evidence for activation of non-canonical NF-kB signaling (Fig. 2c). Next, we generated a dual GFP/luciferase NF-kB reporter Jurkat cell line to measure NF-kB transcriptional activity. Cells transduced with pMSCV-FYN-TRAF3IP2-IRES-mCherry (pMImC-FYN-TRAF3IP2) had increased NF-kB transcriptional activity in resting conditions and after TCR stimulation with an agonistic CD3 antibody compared to Jurkat cells transduced with pMImC-FYN1–232 or pMImC-TRAF3IP2 (Fig. 2d). In contrast, we found no evidence for increased activation of MAPK signaling pathways (Fig. 2e).
Finally, we transduced murine CD4+ T cells to express TRAF3IP2 or FYN-TRAF3IP2 (Supplementary Fig. 3a). Intracellular flow cytometry indicated increased activation of canonical NF-kB signaling by means of increased Inhibitor of NF-kB alpha (IkBa) degradation and increased Ser536 phosphorylation of p65 (Fig. 2f). There was no evidence for enhanced activation of proximal TCR signaling (ZAP70 Tyr318 phosphorylation) or increased MAPK signaling (Fig. 2g).
In summary, these data show that FYN-TRAF3IP2 activates canonical NF-kB signaling independent of IL-17.
FYN-TRAF3IP2 localizes to the cell membrane and activates the NF-kB pathway
Ligation of the TCR engenders the formation of a multiprotein signalosome at the membrane-cytoplasm interface. The Src-family kinases FYN and LCK are critical for proximal TCR signaling and are anchored to the plasma membrane by acylation of the unique N-terminal SH4 domains. FYN is myristoylated at Gly2, followed by palmitoylation at Cys319. IL-17 dependent recruitment of TRAF3IP2 to the plasma membrane initiates signaling downstream of TRAF3IP2. Taken together, we reasoned that acylation of FYN-TRAF3IP2 would anchor the fusion protein to the plasma membrane and mediate chronic active TRAF3IP2-dependent signaling.
To this end, we mutated the N-terminal Gly2 of FYN to Ala (FYNG2A-TRAF3IP2) to abolish acylation of the N-terminal FYN domain. Immunofluorescence imaging of 293T cells transfected with either wild-type TRAF3IP2, FYN-TRAF3IP2 or FYNG2A-TRAF3IP, confirmed that wild-type TRAF3IP2 resided in the cytosol, FYN-TRAF3IP2 segregated to the plasma membrane and FYNG2A-TRAF3IP2 resulted in the redistribution of the fusion protein to the cytosol (Fig. 3a). The subcellular compartmentalization of FYN-TRAF3IP2 and FYNG2A-TRAF3IP2 was confirmed in Ba/F3 cells (Supplementary Fig. 3b) and Ba/F3 cells transduced to express FYNG2A-TRAF3IP2, were no longer able to grow out in the absence of IL-3 (Fig. 3b).
Next, we collected cytosolic and membrane fractions of Jurkat cells with ectopic expression of TRAF3IP2, FYN-TRAF3IP2 or FYNG2A-TRAF3IP2 and confirmed that disruption of FYN myristoylation impeded incorporation of the fusion protein in the plasma membrane (Fig. 3c). Delocalization from the plasma membrane impaired the ability of FYNG2A-TRAF3IP2 to activate NF-kB transcriptional activity compared to FYN-TRAF3IP2 (Fig. 3d). Likewise, immunofluorescence analysis of CD4+ T cells displayed association of FYN-TRAF3IP2 but not TRAF3IP2 or FYNG2A-TRAF3IP2 with the cell membrane (Fig. 3e). Exclusion of FYN-TRAF3IP2 from the membrane compartment, impaired activation of NF-kB signaling in CD4+ T cells (Fig. 3f).
Together, these experiments demonstrate that acylation of the N-terminal SH4 domain of FYN anchors the FYN-TRAF3IP2 fusion protein to the plasma membrane. Partitioning of FYN-TRAF3IP2 to the plasma membrane is required for downstream activation of NF-kB signaling.
FYN-TRAF3IP2 activates TRAF6 independent of the CBM signalosome
Activation of the NF-kB pathway downstream of the TCR is initiated by protein kinase C q (PKCq)-dependent assembly of a signalosome composed of CARD11, BCL10 and MALT1 (CBM signalosome). The CBM signalosome recruits TRAF6, which leads to lysine 63-linked (K63-linked) polyubiquitination of TRAF6. K63-linked polyubiquitinated TRAF6 acts as a scaffold for the IkB Kinase (IKK) complex and Transforming-growth-factor-b-Activated Kinase 1 (TAK1) complex which will ultimately lead to the release of NF-kB transcription factors from IkB20. The TRAF3IP2 protein comprises an N-terminal TRAF6 binding motif21 and has a U-box domain with E3 ubiquitin ligase enzymatic activity through which it catalyzes K63-linked polyubiquitination of TRAF622 (Fig. 1a). We assumed that the interaction of the FYN-TRAF3IP2 fusion protein with TRAF6 would be preserved and enable TRAF6 K63-linked polyubiquitination independent of the CBM signalosome to activate NF-kB.
To test this hypothesis, we substituted the acidic amino acid residues in the N-terminal TRAF6 binding motif (PVEVDE) with Ala residues (PVAVAA). While wild-type TRAF3IP2 and FYN-TRAF3IP2 co-immunoprecipitated with TRAF6, mutation of the TRAF6 binding motif (FYN-TRAF3IP2∆T6) abrogated the interaction with TRAF6 and led to a decrease in K63-linked polyubiquitination of TRAF6 (Fig. 4a). In the opposite direction, co-immunoprecipitation of TRAF6 – including polyubiquitinated TRAF6 – with FYN-TRAF3IP2∆T6 was attenuated (Fig. 4b). Contrary to Ba/F3 cells with expression of FYN-TRAF3IP, Ba/F3 cells with ectopic expression of FYN-TRAF3IP2∆T6 (Supplementary Fig. 3c) did not grow out after withdrawal of IL-3 (Fig. 4c). Expression of FYN-TRAF3IP2∆T6 in Jurkat cells or primary CD4+ T cells (Supplementary Fig. 3c) impaired activation of the canonical NF-kB pathway (Fig. 4d-e).
To investigate whether FYN-TRAF3IP2 could activate the NF-kB pathway independent of CARD11, we generated CARD11 knock-out Jurkat cells with CRISPR/Cas9 genome editing (Supplementary Fig. 3d-e). Expression of FYN-TRAF3IP2, but not TRAF3IP2 or FYN-TRAF3IP2∆T6 augmented NF-kB transcriptional activity in resting conditions in both wild-type and CARD11 knock-out Jurkat cells. Activation of PKCq with phorbol 12-myristate 13-acetate (PMA) and ionomycin increased NF-kB transcriptional activity in wild-type Jurkat cells, but significantly more in Jurkat cells with expression of FYN-TRAF3IP2. CARD11 deficiency impaired NF-kB activation in response to PMA and ionomycin, but CARD11 knock-out Jurkat cells with FYN-TRAF3IP2 expression remained significantly more responsive to PMA/ ionomycin stimulation than empty vector control or cells with expression of TRAF3IP2 or FYN-TRAF3IP2∆T6. This proves that FYN-TRAF3IP2 does not require the CBM signalosome, but suggests an interaction – direct or indirect – between FYN-TRAF3IP2 and PKCq.
Collectively, these results indicate that FYN-TRAF3IP2 directly interacts with TRAF6 and activates TRAF6 and the NF-kB pathway without intervention of the CBM signalosome.
FYN-TRAF3IP2 expression causes PTCL-NOS-like disease in vivo
To test the oncogenic potential of the fusion protein in vivo, we transduced lineage negative hematopoietic stem and progenitor cells (HSPC) from wild-type mice with either the empty pMIG vector or the pMIG vector containing FYN-TRAF3IP2 or FYNG2A-TRAF3IP2. Subsequently, we injected the transduced cells (Supplementary Fig. 4a) intravenously in sublethally irradiated syngeneic recipient mice. Engraftment rates were comparable (Supplementary Fig. 4b). As early as 7 weeks after transplantation, GFP+ cells in the peripheral blood of mice transplanted with HSPC expressing FYN-TRAF3IP2 skewed towards CD4+ cells (Supplementary Fig. 4c). Mice with expression of FYN-TRAF3IP2 in hematopoietic cells, started losing weight 8 weeks after transplantation and succumbed within 16 weeks after transplantation (Fig. 5a). These mice had splenomegaly (Fig. 5b) and generalized lymphadenopathy. In contrast, mice transplanted with cells expressing mutant FYNG2A-TRAF3IP2 did not develop disease (Fig. 5a,b). Lymph nodes from mice with FYN-TRAF3IP2-expressing cells displayed an effaced lymph node architecture with paracortical expansion and loss of germinal centers. The lymph nodes contained medium-sized and large cells with irregular, pleomorphic nuclei and multiple, prominent nucleoli. Malignant cells were surrounded by a polymorphic infiltrate with numerous eosinophils and histiocytes (Fig. 5c). FYN-TRAF3IP2-driven lymphomas were invariably CD4+ (Fig. 5d). These cells were also found in the spleens (Supplementary Fig. 4d) and infiltrated the livers (Fig. 5e-f). FYN-TRAF3IP2 and FYNG2A-TRAF3IP2 were expressed in lymph nodes (Fig. 5g) and FYN-TRAF3IP2 expression was restricted to GFP+ cells (Supplementary Fig. 4e). Analysis of Tcrb locus rearrangements with RT-PCR corroborated the clonal nature of the disease (Fig. 5h).
Because we identified FYN-TRAF3IP2 gene fusions in both PTCL-NOS and PTCL-TFH, we determined the immunophenotype of murine FYN-TRAF3IP2-driven lymphomas. Malignant cells were positive for PD-1 and ICOS, but negative for the Tfh marker CXCR5 (Fig. 6a). The BCL6 transcription factor orchestrates Tfh identity, but was not expressed in FYN-TRAF3IP2-induced lymphomas (Fig. 6b). B cell proliferation with an expansion of germinal center (GC) B cells and plasma cells frequently accompanies Tfh-related neoplasms. In contrast, we observed a reduction in B cells in FYN-TRAF3IP2-associated lymphomas (Fig. 6c) without an increased proportion of GC B cells (Fig. 6d). The presence of plasma cells was more variable, but overall, not significantly altered (Fig. 6e). Consistent with the histologic finding of an increased number of histiocytes and eosinophils, the proportion of CD11b+ myeloid cells was increased in FYN-TRAF3IP2-driven lymphomas (Supplementary Fig. 4f). The fraction of CD4+ cells was increased in FYN-TRAF3IP2 driven lymphomas and there were no changes in the number of CD8+ cells (Supplementary Fig. 4f). Finally, there was no proliferation of high endothelial venules (Fig. 6f). These results are compatible with PTCL-NOS rather than PTCL-TFH.
Collectively, these in vivo data indicate that FYN-TRAF3IP2 is a disease driver in PTCL-NOS and that the oncogenic potential of the FYN-TRAF3IP2 fusion protein is critically dependent on its anchored location to the plasma membrane because lymphomagenesis was abolished in FYNG2A-TRAF3IP2-expressing cells.
FYN-TRAF3IP2-driven lymphomas have active NF-kB signaling and are sensitive to inhibition of BCL-XL
We compared the gene expression profile of sorted CD4+GFP+ lymphoma cells with naive CD4+ T cells, CD4+GFP− non-malignant stromal T cells from the lymphoma and CD4+GFP+ FYNG2A-TRAF3IP2-expressing T cells. Consistent with the neoplastic nature of the disease, numerous cell-cycle-associated genes were among the most significantly upregulated genes in FYN-TRAF3IP2-expressing lymphoma cells (Supplementary Fig. 5a). Overexpression of Icos and Pdcd1 was congruent with the immunophenotype determined with flow cytometry. Additionally, lymphoma cells overexpressed Runx2 and Id2. Both genes oppose the Tfh phenotype and are repressed by Bcl623, in support of a non-Tfh origin of the lymphomas (Supplementary Fig. 5a). We analyzed the cis-regulatory features associated with differentially expressed genes to identify the transcription factors driving these phenotypes with i-cisTarget. This analysis consistently retrieved the NF-kB1 motif and the PU.1 motif as the most significantly enriched cis-regulatory features associated with overexpressed genes in FYN-TRAF3IP2-expressing lymphoma cells (Fig. 7a). Gene set enrichment analysis (GSEA) for different p65 (RelA) target gene gene sets reaffirmed increased expression of canonical NF-kB target genes in FYN-TRAF3IP2-expressing cells (Fig. 7b). As expected, malignant cells expressed Relb – a known direct transcriptional target of p50/p6524 (Supplementary Fig. 5b). However, there was no enrichment of RelB target genes in malignant cells (Supplementary Fig. 5c) and RelB was sequestered in the cytosol of FYN-TRAF3IP2-expressing cells (Supplementary Fig. 5b). The transcription factor PU.1 (also known as SPI1) has been associated with the Th9 phenotype. The cytokine profile of FYN-TRAF3IP2-expressing lymphoma cells was compatible with a Th9 phenotype25 (Supplementary Fig. 5d). Consistent with in vitro assays, there was no univocal activation of MAPK signaling in FYN-TRAF3IP2-expressing cells. The AP-1 motif was associated with cis-regulatory features of upregulated genes in lymphoma cells versus naive CD4+ T cells and downregulated genes in lymphoma cells versus FYNG2A-TRAF3IP2-expressing CD4+ T cells (Fig. 7a). Likewise, GSEA for c-Jun target genes yielded inconsistent results (Supplementary Fig. 5e).
Targeting NF-kB directly for clinical purposes remains elusive because of on-target toxicities. This prompted us to examine vulnerabilities downstream of NF-kB. Promoting cell survival through induction of target genes, is one of the best documented functions of NF-kB26. Indeed, pro-survival and to a lesser extent pro-apoptotic factors were upregulated in FYN-TRAF3IP2-expressing cells (Fig. 8a). We reasoned that inhibition of pro-survival signals could tilt the balance towards apoptosis. BCL-XL (encoded by the Bcl2l1) gene was consistently overexpressed by lymphoma cells at both the RNA level and protein level in our mouse model (Fig. 8a-b). BCL-XL expression was also confirmed in case PTCL2 (Fig. 8c). Compared to naive CD4+ T cells cultured under identical conditions, FYN-TRAF3IP2-expressing cells were > 30-fold more sensitive to inhibition of BCL-XL, BCL-W and BCL2 with ABT-263 treatment (Fig. 8d).
These results demonstrate that FYN-TRAF3IP2 activates NF-kB signaling in vivo and that this confers malignant cells with vulnerability to inhibition of BCL-XL.
KHDRBS1-LCK mediates chronic active TCR signaling
We cloned the open reading frame of the KHDRBS1-LCK fusion transcript derived from patient cDNA in the pMIG vector to study the signaling properties of the KHDRBS1-LCK fusion protein. Here, the kinase domain of LCK was fused to the KHDRBS1 dimerization domain. It thus lacks the nuclear localization signal of KHDRBS1 and the membrane localization motif of LCK (Fig. 1c), suggesting that this protein functions in the cytosol. Indeed, in primary T cells with ectopic expression of KHDRBS1-LCK, the fusion protein accumulated in the cytosol when compared to T cells with ectopic expression of LCK (Fig. 9a). Retroviral transduction of IL-3 dependent Ba/F3 cells with KHDRBS1-LCK (Supplementary Fig. 6a) led to IL-3 independent growth in vitro and this was abolished by an inactivating LCK kinase domain mutation (KHDRBS1-LCKK273R)27 (Fig. 9b). Likewise, inhibition of LCK kinase activity with dasatinib could efficiently block the proliferation of transformed Ba/F3 cells at low nanomolar concentrations (Fig. 9c). KHDRBS1-LCK but not KHDRBS1-LCKK273R was constitutively active (Tyr394 phosphorylation) in unstimulated Jurkat cells (Fig. 9d). Likewise, proximal TCR signaling was significantly enhanced in primary T cells with expression of KHDRSB1-LCK in resting conditions and after TCR stimulation compared to primary T cells transduced with empty pMIG vector or LCK (Fig. 9e). Inhibition of LCK kinase activity with dasatinib reverted these differences (Fig. 9e). Further downstream of the TCR, KHDRBS1-LCK activated MAPK signaling and this was inhibited by dasatinib (Fig. 9f).
Collectively, these data show that KHDRBS1-LCK is a constitutively active tyrosine kinase which leads to chronic active TCR signaling.
KHDRBS1-LCK instigates PTCL-NOS in vivo
To test the oncogenic potential of KHDRBS1-LCK in vivo, we transduced HSPC from wild-type mice with MSCV retrovirus with either an empty pMIG vector or a pMIG vector containing the KHDRBS1-LCK ORF. We observed incomplete disease penetrance (Fig. 10a). One mouse developed a lymphoproliferative disorder with generalized lymphadenopathy, moderate splenomegaly, pleural effusion and thymic enlargement, but no leukocytosis (Supplementary Fig. 6b). Normal tissue architecture was effaced. Residual follicles were pushed to the border of the lymph nodes. The expanded paracortex was dominated by a monotonous proliferation of medium-sized lymphocytes with irregular nuclei with 1 or 2 nucleoli and an increased number of blood vessels (Fig. 10b). Malignant cells were CD4−CD8+ and the majority expressed surface TCRb (Fig. 10c, Supplementary Fig. 6c). Bone marrow infiltration was limited. Negativity for TdT ruled out a lymphoblastic lymphoma/leukemia (Fig. 10d). Lymphomas expressed the KHDRBS1-LCK fusion protein, which was highly phosphorylated (Fig. 10e). Interestingly, also endogenous LCK was highly phosphorylated in lymphomatous spleens compared to spleens from empty pMIG vector mice despite comparable LCK protein levels, hinting at activation of endogenous LCK by KHDRBS1-LCK.