2022
DOI: 10.3389/fonc.2022.877495
|View full text |Cite
|
Sign up to set email alerts
|

p66Shc Deficiency in Chronic Lymphocytic Leukemia Promotes Chemokine Receptor Expression Through the ROS-Dependent Inhibition of NF-κB

Abstract: The microenvironment of lymphoid organs is central to the pathogenesis of chronic lymphocytic leukemia (CLL). Within it, tumor cells find a favourable niche to escape immunosurveillance and acquire pro-survival signals. We have previously reported that a CLL-associated defect in the expression of the pro-apoptotic and pro-oxidant adaptor p66Shc leads to enhanced homing to and accumulation of leukemic cells in the lymphoid microenvironment. The p66Shc deficiency-related impairment in intracellular reactive oxyg… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

1
14
0

Year Published

2023
2023
2024
2024

Publication Types

Select...
5
1

Relationship

3
3

Authors

Journals

citations
Cited by 6 publications
(15 citation statements)
references
References 58 publications
1
14
0
Order By: Relevance
“…p66Shc is a molecular adaptor that promotes apoptosis through an adaptor-independent pro-oxidant activity [21,22], which contributes to the intrinsic apoptosis defects and chemoresistance of CLL cells [17,27,28]. The ROS-elevating, gene-modulating activity of p66Shc also indirectly favours leukemic cell survival by setting an imbalance between the homing and egress receptors that coordinate lymphocyte trafficking, promoting their accumulation in the pro-survival lymphoid niche [50,51]. Here we show that p66Shc deletion in Eμ-TCL1 mice also affects the expression of CLL-critical cytokines such as IL-9 and IL-10 by leukemic cells, which also applies to the p66Shc-deficient CLL cells.…”
Section: Discussionmentioning
confidence: 99%
“…p66Shc is a molecular adaptor that promotes apoptosis through an adaptor-independent pro-oxidant activity [21,22], which contributes to the intrinsic apoptosis defects and chemoresistance of CLL cells [17,27,28]. The ROS-elevating, gene-modulating activity of p66Shc also indirectly favours leukemic cell survival by setting an imbalance between the homing and egress receptors that coordinate lymphocyte trafficking, promoting their accumulation in the pro-survival lymphoid niche [50,51]. Here we show that p66Shc deletion in Eμ-TCL1 mice also affects the expression of CLL-critical cytokines such as IL-9 and IL-10 by leukemic cells, which also applies to the p66Shc-deficient CLL cells.…”
Section: Discussionmentioning
confidence: 99%
“…[114][115][116] The expression of NFR2 target genes, such as NAD (P)H quinone oxidoreductase 1 (NAD(P)H quinone oxidoreductase 1), contributes to a drug-resistant phenotype, whose ablation enhanced the sensitivity of CLL cells to be eliminated by drugs that induce ROS production, such venetoclax. 117 Several evidences support that ROR1 recruits PI3K to induce the transformation of phosphatidylinositol 4,5 -bisphosphate (PIP2) to phosphatidylin- activation also activates mTORC1. p62 binds and increases the activation of mTORC1.…”
Section: Ror1 Role In Drug Resistance Mechanism and Combination Therapymentioning
confidence: 99%
“…In ROR1‐high CLL cells, Wnt5a promotes the expression of NF‐ κ B target genes and thus a significantly greater amount of p62 activating mTORC1 signaling and NRF2 to promote the expression of reactive oxygen species (ROS) detoxifying enzymes thus protecting tumor cells from drug‐induced oxidative stress 114–116 . The expression of NFR2 target genes, such as NAD(P)H quinone oxidoreductase 1 (NAD(P)H quinone oxidoreductase 1), contributes to a drug‐resistant phenotype, whose ablation enhanced the sensitivity of CLL cells to be eliminated by drugs that induce ROS production, such venetoclax 117 . Several evidences support that ROR1 recruits PI3K to induce the transformation of phosphatidylinositol 4,5 ‐bisphosphate (PIP2) to phosphatidylinositol 3–5 ‐trisphosphate (PIP3), leading to the activation of 3‐phosphoinositide‐dependent protein kinase 1, thus to the phosphorylation of AKT and the activation of NF‐ κ B thereby the accumulation of p62 (in a IKKβ‐dependent manner).…”
Section: Ror1 Role In Drug Resistance Mechanism and Combination Therapymentioning
confidence: 99%
“…NF-κB can bind to enhancers and promoters of its target genes, such as matrix metallopeptidase 9 (MMP-9), vascular endothelial growth factor (VEGF), urokinase type plasminogen activator (uPA) and chemokine receptors (CXCR1, CXCR2, CXCR3, CXCR4, CCR2 and CCR7, etc.) that control metastasis, invasion and angiogenesis of cancer cells (Richmond, 2002;Taniguchi and Karin, 2018;Tatangelo et al, 2022). In addition, survival and maturation of B lymphocytes by BAFF/BAFF receptor (BAFFR) are mainly mediated by NF-κB (Zhang et al, 2017).…”
Section: Function and Activation Of Nf-κbmentioning
confidence: 99%