Ikaros sets the threshold for negative B-cell selection by regulation of the signaling strength of the AKT pathway

Standard

Ikaros sets the threshold for negative B-cell selection by regulation of the signaling strength of the AKT pathway. / Ehm, Patrick A H; Horn, Stefan; Hoffer, Konstantin; Kriegs, Malte; Horn, Michael; Giehler, Susanne; Nalaskowski, Marcus; Rehbach, Christoph; Horstmann, Martin A; Jücker, Manfred.

In: CELL COMMUN SIGNAL, Vol. 22, No. 1, 12.07.2024, p. 360.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

APA

Vancouver

Bibtex

@article{eebfa63d36544d848a4dd7e365c858bc,
title = "Ikaros sets the threshold for negative B-cell selection by regulation of the signaling strength of the AKT pathway",
abstract = "Inhibitory phosphatases, such as the inositol-5-phosphatase SHIP1 could potentially contribute to B-cell acute lymphoblastic leukemia (B-ALL) by raising the threshold for activation of the autoimmunity checkpoint, allowing malignant cells with strong oncogenic B-cell receptor signaling to escape negative selection. Here, we show that SHIP1 is differentially expressed across B-ALL subtypes and that high versus low SHIP1 expression is associated with specific B-ALL subgroups. In particular, we found high SHIP1 expression in both, Philadelphia chromosome (Ph)-positive and ETV6-RUNX1-rearranged B-ALL cells. As demonstrated by targeted knockdown of SHIP1 by RNA interference, proliferation of B-ALL cells in vitro and their tumorigenic spread in vivo depended in part on SHIP1 expression. We investigated the regulation of SHIP1, as an important antagonist of the AKT signaling pathway, by the B-cell-specific transcription factor Ikaros. Targeted restoration of Ikaros and pharmacological inhibition of the antagonistic casein kinase 2, led to a strong reduction in SHIP1 expression and at the same time to a significant inhibition of AKT activation and cell growth. Importantly, the tumor suppressive function of Ikaros was enhanced by a SHIP1-dependent additive effect. Furthermore, our study shows that all three AKT isoforms contribute to the pro-mitogenic and anti-apoptotic signaling in B-ALL cells. Conversely, hyperactivation of a single AKT isoform is sufficient to induce negative selection by increased oxidative stress. In summary, our study demonstrates the regulatory function of Ikaros on SHIP1 expression in B-ALL and highlights the relevance of sustained SHIP1 expression to prevent cells with hyperactivated PI3K/AKT/mTOR signaling from undergoing negative selection.",
keywords = "Ikaros Transcription Factor/genetics, Proto-Oncogene Proteins c-akt/metabolism, Signal Transduction, Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics, Humans, B-Lymphocytes/metabolism, Cell Line, Tumor, Cell Proliferation, Animals, Mice",
author = "Ehm, {Patrick A H} and Stefan Horn and Konstantin Hoffer and Malte Kriegs and Michael Horn and Susanne Giehler and Marcus Nalaskowski and Christoph Rehbach and Horstmann, {Martin A} and Manfred J{\"u}cker",
note = "{\textcopyright} 2024. The Author(s).",
year = "2024",
month = jul,
day = "12",
doi = "10.1186/s12964-024-01732-5",
language = "English",
volume = "22",
pages = "360",
journal = "CELL COMMUN SIGNAL",
issn = "1478-811X",
publisher = "BioMed Central Ltd.",
number = "1",

}

RIS

TY - JOUR

T1 - Ikaros sets the threshold for negative B-cell selection by regulation of the signaling strength of the AKT pathway

AU - Ehm, Patrick A H

AU - Horn, Stefan

AU - Hoffer, Konstantin

AU - Kriegs, Malte

AU - Horn, Michael

AU - Giehler, Susanne

AU - Nalaskowski, Marcus

AU - Rehbach, Christoph

AU - Horstmann, Martin A

AU - Jücker, Manfred

N1 - © 2024. The Author(s).

PY - 2024/7/12

Y1 - 2024/7/12

N2 - Inhibitory phosphatases, such as the inositol-5-phosphatase SHIP1 could potentially contribute to B-cell acute lymphoblastic leukemia (B-ALL) by raising the threshold for activation of the autoimmunity checkpoint, allowing malignant cells with strong oncogenic B-cell receptor signaling to escape negative selection. Here, we show that SHIP1 is differentially expressed across B-ALL subtypes and that high versus low SHIP1 expression is associated with specific B-ALL subgroups. In particular, we found high SHIP1 expression in both, Philadelphia chromosome (Ph)-positive and ETV6-RUNX1-rearranged B-ALL cells. As demonstrated by targeted knockdown of SHIP1 by RNA interference, proliferation of B-ALL cells in vitro and their tumorigenic spread in vivo depended in part on SHIP1 expression. We investigated the regulation of SHIP1, as an important antagonist of the AKT signaling pathway, by the B-cell-specific transcription factor Ikaros. Targeted restoration of Ikaros and pharmacological inhibition of the antagonistic casein kinase 2, led to a strong reduction in SHIP1 expression and at the same time to a significant inhibition of AKT activation and cell growth. Importantly, the tumor suppressive function of Ikaros was enhanced by a SHIP1-dependent additive effect. Furthermore, our study shows that all three AKT isoforms contribute to the pro-mitogenic and anti-apoptotic signaling in B-ALL cells. Conversely, hyperactivation of a single AKT isoform is sufficient to induce negative selection by increased oxidative stress. In summary, our study demonstrates the regulatory function of Ikaros on SHIP1 expression in B-ALL and highlights the relevance of sustained SHIP1 expression to prevent cells with hyperactivated PI3K/AKT/mTOR signaling from undergoing negative selection.

AB - Inhibitory phosphatases, such as the inositol-5-phosphatase SHIP1 could potentially contribute to B-cell acute lymphoblastic leukemia (B-ALL) by raising the threshold for activation of the autoimmunity checkpoint, allowing malignant cells with strong oncogenic B-cell receptor signaling to escape negative selection. Here, we show that SHIP1 is differentially expressed across B-ALL subtypes and that high versus low SHIP1 expression is associated with specific B-ALL subgroups. In particular, we found high SHIP1 expression in both, Philadelphia chromosome (Ph)-positive and ETV6-RUNX1-rearranged B-ALL cells. As demonstrated by targeted knockdown of SHIP1 by RNA interference, proliferation of B-ALL cells in vitro and their tumorigenic spread in vivo depended in part on SHIP1 expression. We investigated the regulation of SHIP1, as an important antagonist of the AKT signaling pathway, by the B-cell-specific transcription factor Ikaros. Targeted restoration of Ikaros and pharmacological inhibition of the antagonistic casein kinase 2, led to a strong reduction in SHIP1 expression and at the same time to a significant inhibition of AKT activation and cell growth. Importantly, the tumor suppressive function of Ikaros was enhanced by a SHIP1-dependent additive effect. Furthermore, our study shows that all three AKT isoforms contribute to the pro-mitogenic and anti-apoptotic signaling in B-ALL cells. Conversely, hyperactivation of a single AKT isoform is sufficient to induce negative selection by increased oxidative stress. In summary, our study demonstrates the regulatory function of Ikaros on SHIP1 expression in B-ALL and highlights the relevance of sustained SHIP1 expression to prevent cells with hyperactivated PI3K/AKT/mTOR signaling from undergoing negative selection.

KW - Ikaros Transcription Factor/genetics

KW - Proto-Oncogene Proteins c-akt/metabolism

KW - Signal Transduction

KW - Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics

KW - Humans

KW - B-Lymphocytes/metabolism

KW - Cell Line, Tumor

KW - Cell Proliferation

KW - Animals

KW - Mice

U2 - 10.1186/s12964-024-01732-5

DO - 10.1186/s12964-024-01732-5

M3 - SCORING: Journal article

C2 - 38992657

VL - 22

SP - 360

JO - CELL COMMUN SIGNAL

JF - CELL COMMUN SIGNAL

SN - 1478-811X

IS - 1

ER -