Product Citations: 8

Isoform switch of CD47 provokes macrophage-mediated pyroptosis in ovarian cancer

Preprint on BioRxiv : the Preprint Server for Biology on 18 April 2025 by Wang, Z., Yang, L., et al.

Cancer cells expressing CD47 escape macrophage phagocytosis by binding to the SIRPα ligand expressed on macrophages. CD47 targeted therapy offers a promising approach to cancer treatment. Here we report that two major isoforms of CD47 differ in ovarian cancer and normal tissues. The truncated isoform lacking exon 9 and exon 10 (CD47-S) was exclusively expressed in normal tissues, whereas the full-length isoform (CD47-L) was predominantly expressed in ovarian cancer tissues. Interestingly, CD47-S was unable to locate at the cell surface to bind with SIRPα as CD47-L did, and thereby inactivated “don’t-eat-me” signal. Mechanistic investigations revealed that splicing factor HNRNPA1 promoted splicing switch from CD47-L to CD47-S through exon 9 and exon 10 skipping. We further developed antisense oligonucleotides (ASOs) that effectively switched CD47-L to CD47-S . Importantly, ASOs treatment evoked macrophage-mediated antitumor immune response, thereby triggered pyroptosis of ovarian cancer cells. Moreover, CD47-targeting ASOs significantly reduced tumor growth in patient-derived xenograft. Together, ASO-mediated isoform switch of CD47 has emerged as a promising strategy to improve immune responses against tumors.

  • FC/FACS
  • Cancer Research
  • Immunology and Microbiology

Genetic engineering of allogeneic cell therapeutics that fully prevents rejection by a recipient's immune system would abolish the requirement for immunosuppressive drugs or encapsulation and support large-scale manufacturing of off-the-shelf cell products. Previously, we generated mouse and human hypoimmune pluripotent (HIP) stem cells by depleting HLA class I and II molecules and overexpressing CD47 (B2M-/-CIITA-/-CD47+). To determine whether this strategy is successful in non-human primates, we engineered rhesus macaque HIP cells and transplanted them intramuscularly into four allogeneic rhesus macaques. The HIP cells survived unrestricted for 16 weeks in fully immunocompetent allogeneic recipients and differentiated into several lineages, whereas allogeneic wild-type cells were vigorously rejected. We also differentiated human HIP cells into endocrinologically active pancreatic islet cells and showed that they survived in immunocompetent, allogeneic diabetic humanized mice for 4 weeks and ameliorated diabetes. HIP-edited primary rhesus macaque islets survived for 40 weeks in an allogeneic rhesus macaque recipient without immunosuppression, whereas unedited islets were quickly rejected.
© 2023. The Author(s).

  • FC/FACS
  • Stem Cells and Developmental Biology

Protection of cell therapeutics from antibody-mediated killing by CD64 overexpression.

In Nature Biotechnology on 1 May 2023 by Gravina, A., Tediashvili, G., et al.

Allogeneic cell therapeutics for cancer therapy or regenerative medicine are susceptible to antibody-mediated killing, which diminishes their efficacy. Here we report a strategy to protect cells from antibody-mediated killing that relies on engineered overexpression of the IgG receptor CD64. We show that human and mouse iPSC-derived endothelial cells (iECs) overexpressing CD64 escape antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity from IgG antibodies in vitro and in ADCC-enabled mice. When CD64 expression was combined with hypoimmune genetic modifications known to protect against cellular immunity, B2M-/-CIITA-/- CD47/CD64-transgenic iECs were resistant to both IgG antibody-mediated and cellular immune killing in vitro and in humanized mice. Mechanistic studies demonstrated that CD64 or its intracellularly truncated analog CD64t effectively capture monomeric IgG and occupy their Fc, and the IgG bind and occupy their target antigens. In three applications of the approach, human CD64t-engineered thyroid epithelial cells, pancreatic beta cells and CAR T cells withstood clinically relevant levels of graft-directed antibodies and fully evaded antibody-mediated killing.
© 2023. The Author(s).

  • FC/FACS

Manufacturing autologous chimeric antigen receptor (CAR) T cell therapeutics is complex, and many patients experience treatment delays or cannot be treated at all. Although current allogeneic CAR products have the potential to overcome manufacturing bottlenecks, they are subject to immune rejection and failure to persist in the host, and thus do not provide the same level of efficacy as their autologous counterparts. Here, we aimed to develop universal allogeneic CAR T cells that evade the immune system and produce a durable response. We generated human hypoimmune (HIP) T cells with disrupted B2M, CIITA, and TRAC genes using CRISPR-Cas9 editing. In addition, CD47 and anti-CD19 CAR were expressed using lentiviral transduction. These allogeneic HIP CD19 CAR T cells were compared to allogeneic CD19 CAR T cells that only expressed the anti-CD19 CAR (allo CAR T). In vitro assays for cancer killing and exhaustion revealed no differences between allo CAR T and HIP CAR T cells, confirming that the HIP edits did not negatively affect T cell performance. Clearance of CD19+ tumors by HIP CAR T cells in immunodeficient NSG mice was comparable to that of allo CAR T cells. In fully immunocompetent humanized mice, HIP CAR T cells significantly outperformed allo CAR T cells, showed improved persistence and expansion, and provided lasting cancer clearance. Furthermore, CD47-targeting safety strategies reliably and specifically eliminated HIP CAR T cells. These findings suggest that universal allogeneic HIP CAR T cell-based therapeutics might overcome the limitations associated with poor persistence of allogeneic CAR T cells and exert durable anti-tumor responses.
© 2023. The Author(s).

  • FC/FACS
  • Cancer Research
  • Immunology and Microbiology

Alternative 3' untranslated regions (3' UTRs) are widespread, but their functional roles are largely unknown. We investigated the function of the long BIRC3 3' UTR, which is upregulated in leukemia. The 3' UTR does not regulate BIRC3 protein localization or abundance but is required for CXCR4-mediated B cell migration. We established an experimental pipeline to study the mechanism of regulation and used mass spectrometry to identify BIRC3 protein interactors. In addition to 3'-UTR-independent interactors involved in known BIRC3 functions, we detected interactors that bind only to BIRC3 protein encoded from the mRNA with the long 3' UTR. They regulate several functions, including CXCR4 trafficking. We further identified RNA-binding proteins differentially bound to the alternative 3' UTRs and found that cooperative binding of Staufen and HuR mediates 3'-UTR-dependent complex formation. We show that the long 3' UTR is required for the formation of specific protein complexes that enable additional functions of BIRC3 protein beyond its 3'-UTR-independent functions.
Copyright © 2019 Elsevier Inc. All rights reserved.

  • FC/FACS
  • Homo sapiens (Human)
  • Biochemistry and Molecular biology
View this product on CiteAb