Cyclin D2 Dysregulation by Chromosomal Translocations to TCR Loci in T-Cell Acute Lymphoblastic Leukemia (T-ALL).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2073-2073
Author(s):  
Emmanuelle Clappier ◽  
Wendy Cuccuini ◽  
Jean-Michel Cayuela ◽  
Danielle Vecchione ◽  
Andre Baruchel ◽  
...  

Abstract D-type cyclins are key regulators of progression through G1 phase of the cell cycle. Strong expression of at least one of the three D cyclins is common in human cancers. However, while the cyclin D1 and D3 genes (CCND1 and CCND3) are recurrently involved in genomic rearrangements, especially in mantle-cell lymphoma and multiple myeloma, no clear involvement of the cyclin D2 gene (CCND2) has been reported to date in human malignancies. In T-cell acute lymphoblastic leukemia (T-ALL), the T-cell receptor genes TCRA/D and TCRB are frequently involved in chromosomal rearrangements. In order to identify new genomic rearrangements and oncogenes in human T-ALL, we performed an interphasic FISH screening of T-ALL cases using TCR flanking probes. Using this approach, we identified two new chromosomal translocations: t(7;12)(q34;p13) and t(12;14)(p13;q11), involving the TCRB and TCRA/D loci, respectively. Molecular analysis of the breakpoint sequences demonstrated the involvement of the CCND2 locus at 12p13. Expression analysis using RQ-PCR and immunoblotting demonstrated dramatic cyclin D2 overexpression in the translocated cases (n=3) compared to other T-ALLs (total, n=86), whereas other genes located near the translocation breakpoints were not deregulated on microarray analysis. To further evaluate expression in T-ALL with respect to normal T-cell differentiation, we analyzed CCND2 expression in purified subpopulations from normal human thymus. CCND2 levels were downregulated through progression from the early stages of normal human T-cell differentiation and transition to beta-selection. In the most immature T-ALLs, a moderate CCND2 expression was observed, consistent with their differentiation stage, while low expression was found in other T-ALL. By contrast, the massive and sustained expression in the CCND2-rearranged T-ALL cases strongly suggested an oncogenic role due to the TCR translocation. T-ALL oncogenesis is a multi-step process; we here found that the TCR-CCND2 translocations were associated with other oncogene expression (TAL1, HOXA, or TLX3/HOX11L2), NOTCH1 activating mutations, and/or CDKN2A/p16/ARF deletion, showing that cyclin D2 dysregulation could contribute to multi-event oncogenesis in various T-ALL groups. In conclusion, this report is the first clear evidence of a direct involvement of cyclin D2 in human cancer due to recurrent somatic genetic alterations. This reinforces the view that the strong expression of cyclin D2 which is detected in various types of cancer can contribute to oncogenesis, and points to cyclin D2 as a potential target for therapy in these tumors.

2021 ◽  
Vol 118 (34) ◽  
pp. e2110758118
Author(s):  
Maike Bensberg ◽  
Olof Rundquist ◽  
Aida Selimović ◽  
Cathrine Lagerwall ◽  
Mikael Benson ◽  
...  

Pediatric T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy resulting from overproduction of immature T-cells in the thymus and is typified by widespread alterations in DNA methylation. As survival rates for relapsed T-ALL remain dismal (10 to 25%), development of targeted therapies to prevent relapse is key to improving prognosis. Whereas mutations in the DNA demethylating enzyme TET2 are frequent in adult T-cell malignancies, TET2 mutations in T-ALL are rare. Here, we analyzed RNA-sequencing data of 321 primary T-ALLs, 20 T-ALL cell lines, and 25 normal human tissues, revealing that TET2 is transcriptionally repressed or silenced in 71% and 17% of T-ALL, respectively. Furthermore, we show that TET2 silencing is often associated with hypermethylation of the TET2 promoter in primary T-ALL. Importantly, treatment with the DNA demethylating agent, 5-azacytidine (5-aza), was significantly more toxic to TET2-silenced T-ALL cells and resulted in stable re-expression of the TET2 gene. Additionally, 5-aza led to up-regulation of methylated genes and human endogenous retroviruses (HERVs), which was further enhanced by the addition of physiological levels of vitamin C, a potent enhancer of TET activity. Together, our results clearly identify 5-aza as a potential targeted therapy for TET2-silenced T-ALL.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 474-478 ◽  
Author(s):  
LM Weiss ◽  
JM Bindl ◽  
VJ Picozzi ◽  
MP Link ◽  
RA Warnke

A series of 26 lymphoblastic lymphomas (LLs) and 13 T cell acute lymphoblastic leukemias (ALLs) were investigated using a battery of monoclonal antibodies applied to tissue frozen sections. Twenty-one of the LLs were of T lineage. All but one of the T cell LLs were of immature thymic phenotype, mostly corresponding to stage II cortical thymocyte development. The T cell LLs expressed Leu-1 in 100%, Leu-4 and Leu-9 in 95%, and Leu-5 in 85% of the cases. The high percentage of Leu-4 expression in this series is probably due to detection of cytoplasmic antigen with our methods. One LL was of pre-B or B cell and two cases were of common ALL phenotype. Two cases were of undefined phenotype, expressing markers of both B and T cell differentiation. Pediatric cases showed a greater tendency toward T cell phenotype than did adult cases. The cases of T cell ALL were immunophenotypically similar to the cases of T cell LL but showed a tendency toward a more immature phenotype.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1409-1409
Author(s):  
Eric M Vroegindeweij ◽  
Tim Lammens ◽  
Erik Splinter ◽  
Anne Uyttebroeck ◽  
Kirsten Canté-Barrett ◽  
...  

Abstract Background: T-cell acute lymphoblastic leukemia is characterized by clonal and mutual exclusive chromosomal rearrangements that recurrently activate TAL1, LMO2, TLX1, NKX2-1, TLX3, HOXA or MEF2C oncogenes. Most of these translocations or chromosomal rearrangements occur as erroneous D-J or V-DJ rearrangement attempts of T-cell receptor beta (TCRB) or TCR alpha/delta (TCRAD) genes, mostly positioning oncogenes under the transcriptional control of TCR enhancer elements. Alternatively, oncogenes can also be activated as consequence of BCL11B chromosomal rearrangements. Although many oncogenes are known in T-ALL, the driving oncogenic lesion in particular T-ALL cases remains unknown. Aims: In this study, we aimed to clone reciprocal breakpoint sequences to elucidate cellular mechanisms that lead to recurrent BCL11B -TLX3 chromosomal translocations. Moreover, we want to identify oncogene candidates in various T-ALL patient samples with BCL11B-, TCRB- or TCRAD-translocations for which the candidate oncogene so far has not been identified. Methods: We used Targeted Locus Amplification procedure, a recently developed method that relies on the crosslinking of DNA in live cells, DNA digestion and re-ligation to allow formation of circular DNA ligation fragments and inverted polymerase chain reaction amplification from specific view-point loci. Amplified DNA fragments are sequenced by next generation sequencing, allowing sequence identification in a region covering 2MB around selected regions of interest, including TLX3, TLX1, TAL1, LMO2, BCL11B, TCRAD (TRAJ61), TCRB (TCRBC2) Results: TLA was successfully performed on 10 T-ALL patients having FISH validated TAL1 translocations (2 patients), LMO2 translocations (3 patients), TLX3 translocations (3 patients), TLX1 translocations (2 patients) or an inversion targeting NKX2.1 (1 patient). Analysis of both TAL1 translocated cases revealed a TAL1-TCRAD genomic fusion due to a classical t(1;14)(q32;q11) in 1 patient, but surprisingly reveal a TAL1-TCF7 genomic fusion due to a t(1;5)(q32;q31.1) chromosomal translocation in the second patient. For the LMO2 translocated cases, two patients showed classical LMO2-TCRAD (t(11;14)(p13;q11)) or LMO2-TCRB (t(7;11)(q35;p13) translocations, whereas the third patient presented with an unusual LMO2-BCL11B genomic fusion due to a t(11;14)(p13;q32). Two out of 3 TLX3-translocated patients had classical t(5;14)(q35;q32) translocations, whereas the TLX3 gene in the third patient was rearranged to the calcyphosine-like gene (CAPSL, which flanks the IL7Ra gene) on chromosomal 5p13.2 due to a t(5;5)( p13.2;q35) or an inv(5). One patient had an inversion on chromosome 14, i.e. inv(14)(q11;q13), that brings the NKX2.1 oncogene under the transcriptional control of the TCRAD enhancer. Finally, one TLX1-rearranged patient had a classical TLX1-TCRAD translocation, whereas the other presented with a chromosomal inversion involving the chromosomal band 10q24 (which included TLX1), but also revealed a novel translocation involving the centromere protein P gene (CENPP) on chromosome 9q22.31 with the TCRAD locus. Summary/Conclusions: Targeted Locus Amplification identification of chromosomal rearrangements and genomic breakpoint sequences reveals novel complex translocations in 3 out of 10 T-ALL cases analyzed thus far, indicating higher complexity of chromosomal translocations of known T-ALL oncogenes as thus far anticipated. It further proved a useful tool to identify novel translocation partners from various loci such as the TCR or BCL11B genes that are recurrently involved in these chromosomal rearrangements in T-ALL. Cloning of molecular translocation breakpoints of diagnostic T-ALL patient samples may further provide excellent minimal residual disease markers for disease monitoring during the course of treatment. Disclosures Splinter: Cergentis BV: Employment. van Min:Cergentis BV: Employment.


Blood ◽  
2017 ◽  
Vol 130 (15) ◽  
pp. 1722-1733 ◽  
Author(s):  
AHyun Choi ◽  
Anuradha Illendula ◽  
John A. Pulikkan ◽  
Justine E. Roderick ◽  
Jessica Tesell ◽  
...  

Key Points RUNX1 maintains Myb and Myc enhancer activity and is required for leukemogenesis in vivo. RUNX1 inhibition impairs the growth of primary T-ALL patient cells without an effect on normal human hematopoietic cells.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 474-478 ◽  
Author(s):  
LM Weiss ◽  
JM Bindl ◽  
VJ Picozzi ◽  
MP Link ◽  
RA Warnke

Abstract A series of 26 lymphoblastic lymphomas (LLs) and 13 T cell acute lymphoblastic leukemias (ALLs) were investigated using a battery of monoclonal antibodies applied to tissue frozen sections. Twenty-one of the LLs were of T lineage. All but one of the T cell LLs were of immature thymic phenotype, mostly corresponding to stage II cortical thymocyte development. The T cell LLs expressed Leu-1 in 100%, Leu-4 and Leu-9 in 95%, and Leu-5 in 85% of the cases. The high percentage of Leu-4 expression in this series is probably due to detection of cytoplasmic antigen with our methods. One LL was of pre-B or B cell and two cases were of common ALL phenotype. Two cases were of undefined phenotype, expressing markers of both B and T cell differentiation. Pediatric cases showed a greater tendency toward T cell phenotype than did adult cases. The cases of T cell ALL were immunophenotypically similar to the cases of T cell LL but showed a tendency toward a more immature phenotype.


Sign in / Sign up

Export Citation Format

Share Document