Abstract 2469: The B-cell lymphoma specific aptamer C10.36 binds a ribonucleoprotein complex on the cell surface of cancer cells

Author(s):  
Sonal S. Tonapi ◽  
Janet E. Duncan ◽  
Matthew Rosenow ◽  
Melissa Richards ◽  
Teresa L. Tinder ◽  
...  
2020 ◽  
Vol 20 (4) ◽  
pp. 504-517
Author(s):  
Yu-Lan Li ◽  
Xin-Li Gan ◽  
Rong-Ping Zhu ◽  
Xuehong Wang ◽  
Duan-Fang Liao ◽  
...  

Objective: To overcome the disadvantages of cisplatin, numerous platinum (Pt) complexes have been prepared. However, the anticancer activity and mechanism of Pt(II) complexed with 2-benzoylpyridine [Pt(II)- Bpy]: [PtCl2(DMSO)L] (DMSO = dimethyl sulfoxide, L = 2-benzoylpyridine) in cancer cells remain unknown. Methods: Pt(II)-Bpy was synthesized and characterized by spectrum analysis. Its anticancer activity and underlying mechanisms were demonstrated at the cellular, molecular, and in vivo levels. Results: Pt(II)-Bpy inhibited tumor cell growth, especially HepG2 human liver cancer cells, with a halfmaximal inhibitory concentration of 9.8±0.5μM, but with low toxicity in HL-7702 normal liver cells. Pt(II)- Bpy induced DNA damage, which was demonstrated through a marked increase in the expression of cleavedpoly (ADP ribose) polymerase (PARP) and gamma-H2A histone family member X and a decrease in PARP expression. The interaction of Pt(II)-Bpy with DNA at the molecular level was most likely through an intercalation mechanism, which might be evidence of DNA damage. Pt(II)-Bpy initiated cell cycle arrest at the S phase in HepG2 cells. It also caused severe loss of the mitochondrial membrane potential; a decrease in the expression of caspase-9 and caspase-3; an increase in reactive oxygen species levels; the release of cytochrome c and apoptotic protease activation factor; and the activation of caspase-9 and caspase-3 in HepG2 cells, which in turn resulted in apoptosis. Meanwhile, changes in p53 and related proteins were observed including the upregulation of p53, the phosphorylation of p53, p21, B-cell lymphoma-2-associated X protein, and NOXA; and the downregulation of B-cell lymphoma 2. Moreover, Pt(II)-Bpy displayed marked inhibitory effects on tumor growth in the HepG2 nude mouse model. Conclusion: Pt(II)-Bpy is a potential candidate for cancer chemotherapy.


2011 ◽  
Vol 130 (8) ◽  
pp. 1755-1767 ◽  
Author(s):  
Alexandre Rouette ◽  
Sophie Parent ◽  
Julie Girouard ◽  
Valérie Leblanc ◽  
Eric Asselin

2020 ◽  
Author(s):  
Abdessamad Zerrouqi ◽  
Anna Torun ◽  
Nina Miazek ◽  
Zofia Pilch ◽  
Jakub Golab ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1692-1692 ◽  
Author(s):  
Marco Fangazio ◽  
David Dominguez-Sola ◽  
Fabrizio Tabbò ◽  
Davide Rossi ◽  
Julie Teruya-Feldstein ◽  
...  

Abstract Diffuse large B cell lymphoma (DLBCL) is the most common form of B cell non-Hodgkin lymphoma (B-NHL), accounting for ~25-40% of all lymphoid tumors. DLBCL comprises genetically, phenotypically and clinically distinct subtypes, including the prognostically favorable germinal center B cell like (GCB)-DLBCL and the more aggressive activated B cell like (ABC)-DLBCL. We have shown that >60% of DLBCL, independent of molecular subtype, lack cell surface expression of HLA-class I (HLA-I), suggesting that these tumors may escape immune recognition by cytotoxic T cells (CTL) (Challa-Malladi, Lieu et al., Cancer Cell, 2011). HLA-I loss also represents a common lesion acquired at transformation of follicular lymphoma (FL) to DLBCL (Pasqualucci et al., Cell Reports 2014). We have investigated the expression of HLA-I across the clinico-pathological spectrum of mature B cell tumors, and found that HLA-I loss is significantly less common in other mature B-NHL, including Burkitt lymphoma (13/43, 30.2%; p=.002), FL (12/60, 20.0%; p<.001), mantle cell lymphoma (1/38, 2.6%; p<.001), marginal zone lymphoma (0/39, 0%; p<.001), and chronic lymphocytic leukemia (1/36, 2.8%; p<.001). These results suggest that HLA-I loss and, thus, escape from recognition from CTL is an important pathogenetic feature of DLBCL. One mechanism of HLA-I loss, identified by exome-sequencing and copy number analysis, is represented by genomic deletions and/or mutational inactivation of the B2M gene, which are found in ~50% of HLA-I negative cases (29% of all DLBCL). These lesions lead to the complete loss of B2-microglobulin, a required component for the assembly and cell surface expression of the HLA-I complex (Pasqualucci et al. Nat Genet, 2011; Challa-Malladi, Lieu et al. Cancer Cell, 2011). However, the remaining ~50% of patients lack surface HLA-I despite the absence of B2M genetic lesions, suggesting the existence of additional underlying mechanisms. In particular, a fraction of patients express an intact B2M protein, which is mislocalized to the cytoplasm. To investigate whether direct genetic disruption of the HLA-I genes could be responsible for the lack of surface HLA-I in these cases, we performed Sanger sequencing and SNP6.0 array analysis of the HLA-I heavy chain genes (HLA-A and HLA-B) in two DLBCL cell lines (Ly10 and RCK8) with wild-type B2M alleles, but cytoplasmic B2M protein. In both lines, we found the presence of biallelic mutations or deletions in the HLA-I loci. Accordingly, transduction with a retrovirus expressing either HLA-I gene was sufficient to restore cell surface B2M and HLA-I in both lines, documenting that DLBCL can exploit genetic disruption of HLA-I as an alternative mechanism to impair the assembly of a membrane HLA-I complex. The overall contribution of this mechanism to HLA-I loss is currently being determined by using a custom capture/next generation sequencing approach of the HLA-I loci in a large panel of paired tumor/normal biopsies with negative or mislocalized B2M/HLA-I. We also examined the role of B2M (HLA-I) loss in lymphomagenesis in vivo. Particularly, since constitutional B2m deletion is not tumorigenic per se (Koller et al., Science 1990), and B2M loss is frequently acquired during FL transformation to DLBCL, we investigated whether the absence of major histocompatibility complex on the cell surface of mature B cells accelerates tumorigenesis in the presence of other oncogenic lesions. To this end, we generated a conditional knock-out mouse model in which the B2m gene is specifically deleted in germinal center B cells upon expression of a Cγ1-Cre allele, and crossed them with IµHABCL6 knock-in mice, which develop DLBCL due to deregulated expression of the BCL6 oncogene (Cattoretti, Pasqualucci et al., Cancer Cell 2006). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (23) ◽  
pp. 4635-4644 ◽  
Author(s):  
Mary C. Clark ◽  
Mabel Pang ◽  
Daniel K. Hsu ◽  
Fu-Tong Liu ◽  
Sven de Vos ◽  
...  

Abstract Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma and an aggressive malignancy. Galectin-3 (gal-3), the only antiapoptotic member of the galectin family, is overexpressed in DLBCL. While gal-3 can localize to intracellular sites, gal-3 is secreted by DLBCL cells and binds back to the cell surface in a carbohydrate-dependent manner. The major counterreceptor for gal-3 on DLBCL cells was identified as the transmembrane tyrosine phosphatase CD45. Removal of cell-surface gal-3 from CD45 with the polyvalent glycan inhibitor GCS-100 rendered DLBCL cells susceptible to chemotherapeutic agents. Binding of gal-3 to CD45 modulated tyrosine phosphatase activity; removal of endogenous cell-surface gal-3 from CD45 with GCS-100 increased phosphatase activity, while addition of exogenous gal-3 reduced phosphatase activity. Moreover, the increased susceptibility of DLBCL cells to chemotherapeutic agents after removal of gal-3 by GCS-100 required CD45 phosphatase activity. Gal-3 binding to a subset of highly glycosylated CD45 glycoforms was regulated by the C2GnT-1 glycosyltransferase, indicating that specific glycosylation of CD45 is important for regulation of gal-3–mediated signaling. These data identify a novel role for cell-surface gal-3 and CD45 in DLBCL survival and suggest novel therapeutic targets to sensitize DLBCL cells to death.


2020 ◽  
Vol 17 (8) ◽  
pp. 943-953
Author(s):  
Zhe Yin ◽  
Donglin Yang ◽  
Jun Wang ◽  
Yuequan Jiang

Proteins of B-cell lymphoma (Bcl-2) family are key regulators of apoptosis and are involved in the pathogenesis of various cancers. Disrupting the interactions between the antiapoptotic and proapoptotic Bcl-2 members is an attractive strategy to reactivate the apoptosis of cancer cells. Structure-based drug design (SBDD) has been successfully applied to the discovery of small molecule inhibitors targeting Bcl-2 proteins in past decades. Up to now, many Bcl-2 inhibitors with different paralogue selectivity profiles have been developed and some were used in clinical trials. This review focused on the recent applications of SBDD strategies in the development of small molecule inhibitors targeting Bcl-2 family proteins.


BMC Cancer ◽  
2014 ◽  
Vol 14 (1) ◽  
Author(s):  
Qiang Wu ◽  
Xue Liu ◽  
Hong Yan ◽  
Yin-huan He ◽  
Shan Ye ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document