CD7/CD19 Double-Positive T-Cell Acute Lymphoblastic Leukemia

2006 ◽  
Vol 83 (4) ◽  
pp. 324-327
Author(s):  
Shinya Fujisawa ◽  
Fumihiko Tanioka ◽  
Toshihiko Matsuoka ◽  
Takachika Ozawa ◽  
Kensuke Naito ◽  
...  
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 143-143
Author(s):  
Emmanuel Volanakis ◽  
Richard T Williams ◽  
Charles J. Sherr

Abstract Abstract 143 At presentation, 50% of T cell acute lymphoblastic leukemia (T ALL) cases harbor activating mutations of the NOTCH1 transmembrane receptor, virtually all of which have also sustained deletions of the CDKN2A (INK4A-ARF) locus. Although the concordance of these genetic anomalies has been well documented, its basis remains unclear. The CDKN2A gene cluster encodes two structurally and functionally distinct tumor suppressors, p16INK4A and p14ARF (p19Arf in the mouse), the activation of which modulates the activities of the retinoblastoma protein and p53, respectively, to induce cell cycle arrest and/or apoptosis in response to oncogenic stress. A novel approach for modeling T ALL in the mouse now reveals that the ability of Arf to suppress Notch1-induced tumors is conferred at a specific stage of T cell development. Bone marrow cells or thymocytes transduced with a vector encoding the constitutively active intracellular fragment of NOTCH1 (ICN1) together with green fluorescent protein (GFP) were cultured ex vivo under conditions that support T lymphocyte differentiation and proliferation. These cultures were quickly dominated by early T cell precursors that expressed Thy-1 but not B cell or myeloid markers, and which produced a rapidly fatal CD4+/CD8+ (”double-positive”) T ALL when transferred into healthy, non-irradiated syngeneic mice. A phenotypically identical disease resulted upon infusion of the transduced cells into athymic nude mice, demonstrating that the deregulated ICN1 signal is sufficient to drive T cell development to the CD4+/CD8+ stage without any requirement for input from the thymic microenvironment. In recipients of bone marrow-derived ICN1+, Arf+/+ progenitors, T ALLs arose at high frequency, retained the Arf locus, but universally failed to express p19Arf. In turn, T ALLs initiated with bone marrow-derived ICN1+, Arf-/- donor cells exhibited only a modest acceleration of disease progression. In marked contrast, retention of the intact Arf locus in thymocyte-derived ICN1+ donor cells significantly prolonged disease latency and decreased tumor penetrance, indicating that Arf tumor suppression can be activated in thymic progenitors, but not in their less mature bone marrow precursors. Polycomb complexes epigenetically silence the Ink4a-Arf locus in primitive hematopoietic progenitors and early T cell precursors, preventing p19Arf expression and licensing a robust proliferative response to ICN1 signals. However, the locus is subsequently remodeled before T cells reach the double-positive stage, and its activation enables the Arf-p53 axis to cull T cells exposed to aberrant ICN1-induced oncogenic signals. In order to directly demonstrate that ICN1 overexpression in thymocyte-derived progenitors can induce the Arf locus, thymocytes from a homozygous Arf-GFP “knock-in” mouse were transduced with a vector encoding ICN1 and cherry-fluorescent protein (CFP), cultured short-term, and infused into healthy recipient mice. Notably, the Arf-GFP allele, while functionally null, expresses GFP in lieu of p19Arf when the cellular Arf promoter is induced. Healthy syngeneic recipients of ICN1/CFP, ArfGfp/Gfp donor cells developed CFP-marked T ALLs that co-expressed GFP, providing direct evidence that Arf is induced during thymus-derived leukemogenesis in vivo. Notably, Arf-induction not only provides p53-dependent elimination of incipient tumor cells but also generates a selective pressure for the emergence and survival of rare clones that have sustained Arf deletions. We reason that deletion of INK4A-ARF observed in human NOTCH1-induced CD4+/CD8+ T ALL strongly argues for the obligate induction of the locus at an earlier stage of T cell maturation. Our mouse model recapitulates this requirement, and should provide a useful platform to further elucidate disease mechanisms and potential therapies in T ALLs arising in nonirradiated mice that retain hematopoietic and immune functions. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Kehan Li ◽  
Cunte Chen ◽  
Rili Gao ◽  
Xibao Yu ◽  
Youxue Huang ◽  
...  

AbstractT-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of leukemia with poor prognosis, and biomarkers and novel therapeutic targets are urgently needed for this disease. Our previous studies have found that inhibition of the B-cell leukemia/lymphoma 11B (BCL11B) gene could significantly promote the apoptosis and growth retardation of T-ALL cells, but the molecular mechanism underlying this effect remains unclear. This study intends to investigate genes downstream of BCL11B and further explore its function in T-ALL cells. We found that PTK7 was a potential downstream target of BCL11B in T-ALL. Compared with the healthy individuals (HIs), PTK7 was overexpressed in T-ALL cells, and BCL11B expression was positively correlated with PTK7 expression. Importantly, BCL11B knockdown reduced PTK7 expression in T-ALL cells. Similar to the effects of BCL11B silencing, downregulation of PTK7 inhibited cell proliferation and induced apoptosis in Molt-4 cells via up-regulating the expression of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p27. Altogether, our studies suggest that PTK7 is a potential downstream target of BCL11B, and downregulation of PTK7 expression via inhibition of the BCL11B pathway induces growth retardation and apoptosis in T-ALL cells.


2015 ◽  
Vol 208 (1-2) ◽  
pp. 52-53 ◽  
Author(s):  
Xiaolin Ma ◽  
Lijun Wen ◽  
Lili Wu ◽  
Qingrong Wang ◽  
Hong Yao ◽  
...  

2021 ◽  
Vol 22 (3) ◽  
pp. 1388
Author(s):  
Natalia Maćkowska ◽  
Monika Drobna-Śledzińska ◽  
Michał Witt ◽  
Małgorzata Dawidowska

Distinct DNA methylation signatures, related to different prognosis, have been observed across many cancers, including T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological neoplasm. By global methylation analysis, two major phenotypes might be observed in T-ALL: hypermethylation related to better outcome and hypomethylation, which is a candidate marker of poor prognosis. Moreover, DNA methylation holds more than a clinical meaning. It reflects the replicative history of leukemic cells and most likely different mechanisms underlying leukemia development in these T-ALL subtypes. The elucidation of the mechanisms and aberrations specific to (epi-)genomic subtypes might pave the way towards predictive diagnostics and precision medicine in T-ALL. We present the current state of knowledge on the role of DNA methylation in T-ALL. We describe the involvement of DNA methylation in normal hematopoiesis and T-cell development, focusing on epigenetic aberrations contributing to this leukemia. We further review the research investigating distinct methylation phenotypes in T-ALL, related to different outcomes, pointing to the most recent research aimed to unravel the biological mechanisms behind differential methylation. We highlight how technological advancements facilitated broadening the perspective of the investigation into DNA methylation and how this has changed our understanding of the roles of this epigenetic modification in T-ALL.


Sign in / Sign up

Export Citation Format

Share Document