scholarly journals Insights into the mechanism of cell death induced by saporin delivered into cancer cells by an antibody fusion protein targeting the transferrin receptor 1

2013 ◽  
Vol 27 (1) ◽  
pp. 220-231 ◽  
Author(s):  
Tracy R. Daniels-Wells ◽  
Gustavo Helguera ◽  
José A. Rodríguez ◽  
Lai Sum Leoh ◽  
Michael A. Erb ◽  
...  
Blood ◽  
2006 ◽  
Vol 108 (8) ◽  
pp. 2745-2754 ◽  
Author(s):  
Patrick P. Ng ◽  
Gustavo Helguera ◽  
Tracy R. Daniels ◽  
Simon Z. Lomas ◽  
Jose A. Rodriguez ◽  
...  

AbstractWe have previously reported that an anti-human transferrin receptor IgG3-avidin fusion protein (anti-hTfR IgG3-Av) inhibits the proliferation of an erythroleukemia-cell line. We have now found that anti-hTfR IgG3-Av also inhibits the proliferation of additional human malignant B and plasma cells. Anti-hTfR IgG3-Av induces internalization and rapid degradation of the TfR. These events can be reproduced in cells treated with anti-hTfR IgG3 cross-linked with a secondary Ab, suggesting that they result from increased TfR cross-linking. Confocal microscopy of cells treated with anti-hTfR IgG3-Av shows that the TfR is directed to an intracellular compartment expressing the lysosomal marker LAMP-1. The degradation of TfR is partially blocked by cysteine protease inhibitors. Furthermore, cells treated with anti-hTfR IgG3-Av exhibit mitochondrial depolarization and activation of caspases 9, 8, and 3. The mitochondrial damage and cell death can be prevented by iron supplementation, but cannot be fully blocked by a pan-caspase inhibitor. These results suggest that anti-hTfR IgG3-Av induces lethal iron deprivation, but the resulting cell death does not solely depend on caspase activation. This report provides insights into the mechanism of cell death induced by anti-TfR Abs such as anti-hTfR IgG3-Av, a molecule that may be useful in the treatment of B-cell malignancies such as multiple myeloma.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1608-1608
Author(s):  
Jose A Rodriguez ◽  
Gustavo Helguera ◽  
Tracy R Daniels ◽  
David Casero Diaz-Cano ◽  
Elizabeth Ortiz-Sanchez ◽  
...  

Abstract Transferrin receptor 1 (TfR1 or CD71) is the major gateway for iron entry into cells and is overexpressed on a variety of cancer cells. Antibodies targeting TfR1 have shown efficacy in vitro and in vivo against different malignancies by affecting internalization of iron into target cells; however, in some cases their response rate is limited despite the high expression of TfR1 on the target cells. The cause of this limited response is still not well understood. We have developed a mouse/human chimeric IgG3-avidin fusion protein (ch128.1Av) that binds specifically to human TfR1 and exhibits a significant intrinsic anti-proliferative and pro-apoptotic activity against hematopoietic malignant cells by inducing TfR1 degradation and iron starvation. We have previously found a wide range of responses to treatment with ch128.1Av, which can be categorized as high, intermediate, or low sensitivity to the drug. Among the cells tested, the human B-lymphoblastoid cell line IM-9 was one of the most sensitive to ch128.1Av, while the human myeloma cell line U266 was tolerant. To identify the mechanisms responsible for mediating sensitivity or tolerance to the treatment with ch128.1Av, we conducted confocal microscopy studies and a whole genome time-course microarray analysis of IM-9 and U266 cells treated with this drug. In the current work we describe the molecular pathways associated with sensitivity or resistance to this therapy in vitro. We demonstrate that IM-9 cells treated with ch128.1Av can re-route TfR into the lysosomal compartment and exhibit a cytotoxic response that is predicted to be mediated in part by p53 and its target genes based on a transcription factor binding site analysis of the promoter region of genes affected by treatment with ch128.1Av. We show that a siRNA mediated reduction of wild type p53 expression rescues in part the inhibition of proliferation of IM-9 cells treated with ch128.1Av, which validates the in silico promoter analysis. In contrast, we show that U266 cells are tolerant to the cytotoxic response induced by ch128.1Av, probably due to a combination of factors including their failure to redirect trafficking of TfR targeted by ch128.1Av into the lysosomal compartment, lack of a wild type copy of the p53 protein, an early pro-proliferative response to treatment, and a high expression of the cyclin D1 gene. This work has important implications in the clinical use of TfR targeted therapies and is broadly applicable to the advancement and development of novel therapeutics targeting iron metabolism in hematopoietic malignancies. Figure Figure Hierarchical clustering of expression changes (p<0.05) in ch128.1Av treated IM-9 and U266 cells, or between the two cell lines sampled at 1 h, 3 h, 9 h, and 24 h post treatment. The hierarchical clustering of genes and treatment times is based on their expression changes and determined by an un-centered correlation algorithm. Genes shown are also those with the greatest time-dependent variance in ch128.1Av treated IM-9 and U266 cells compared to their time-matched controls. Color scale-bar included shows fold changes in gene expression in log base 2 scale.


PLoS ONE ◽  
2012 ◽  
Vol 7 (9) ◽  
pp. e44482 ◽  
Author(s):  
Tobias Jahn ◽  
Martin Zuther ◽  
Björn Friedrichs ◽  
Claudia Heuser ◽  
Stefan Guhlke ◽  
...  

2019 ◽  
Vol 17 (1) ◽  
pp. 360-360 ◽  
Author(s):  
Rudy Chang ◽  
Abrar Al Maghribi ◽  
Victoria Vanderpoel ◽  
Vitaly Vasilevko ◽  
David H. Cribbs ◽  
...  

2011 ◽  
Vol 286 (41) ◽  
pp. 35708-35715 ◽  
Author(s):  
Jinlong Jian ◽  
Qing Yang ◽  
Xi Huang

Transferrin receptor 1 (TfR1) is a ubiquitous type II membrane receptor with 61 amino acids in the N-terminal cytoplasmic region. TfR1 is highly expressed in cancer cells, particularly under iron deficient conditions. Overexpression of TfR1 is thought to meet the increased requirement of iron uptake necessary for cell growth. In the present study, we used transferrin (Tf), a known ligand of TfR1, and gambogic acid (GA), an apoptosis-inducing agent and newly identified TfR1 ligand to investigate the signaling role of TfR1 in breast cancer cells. We found that GA but not Tf induced apoptosis in a TfR1-dependent manner in breast cancer MDA-MB-231 cells. Estrogen receptor-positive MCF-7 cells lack caspase-3 and were not responsive to GA treatment. GA activated the three major signaling pathways of the MAPK family, as well as caspase-3, -8, and Poly(ADP-ribose)polymerase apoptotic pathway. Interestingly, only Src inhibitor PP2 greatly sensitized the cells to GA-mediated apoptosis. Further investigations by confocal fluorescence microscopy and immunoprecipitation revealed that Src and TfR1 are constitutively bound. Using TfR1-deficient CHO TRVB cells, point mutation studies showed that Tyr20 within the 20YTRF23 motif of the cytoplasmic region of TfR1 is the phosphorylation site by Src. TfR1 Tyr20 phosphomutants were more sensitive to GA-mediated apoptosis. Our results indicate that, albeit its iron uptake function, TfR1 is a signaling molecule and tyrosine phosphorylation at position 20 by Src enhances anti-apoptosis and potentiates breast cancer cell survival.


Sign in / Sign up

Export Citation Format

Share Document