scholarly journals The SPPL3-defined glycosphingolipid repertoire regulates immune responses by improving HLA class I access

2020 ◽  
Author(s):  
Marlieke L.M. Jongsma ◽  
Matthijs Raaben ◽  
Antonius A. de Waard ◽  
Tao Zhang ◽  
Birol Cabukusta ◽  
...  

SummaryHLA class I (HLA-I) drives immune responses by presenting antigen-derived peptides to cognate CD8+ T cells. This process is often hijacked by tumors and pathogens for immune evasion. Since therapeutic options for restoring HLA-I antigen presentation are limited, we aimed to identify new HLA-I pathway targets. By iterative genome-wide screens we uncovered that the cell surface glycosphingolipid (GSL) repertoire determines effective HLA-I antigen presentation. We show that absence of the protease SPPL3 augments B3GNT5 enzyme activity, resulting in upregulated levels of surface (neo)lacto-series GSLs. These GSLs sterically impede molecular interactions with HLA-I and diminish CD8+ T cell activation. In accordance, a disturbed SPPL3-B3GNT5 pathway in glioma associates with decreased patient survival. Importantly, we show that this immunomodulatory effect can be reversed through GSL synthesis inhibition using clinically approved drugs. Overall, our study identifies a GSL signature that functionally inhibits antigen presentation and represents a potential therapeutic target in cancer, infection and autoimmunity.

2007 ◽  
Vol 180 (1) ◽  
pp. 372-382 ◽  
Author(s):  
Andreas Goldwich ◽  
Sabine S. C. Hahn ◽  
Sandra Schreiber ◽  
Stefanie Meier ◽  
Eckhart Kämpgen ◽  
...  

1987 ◽  
Vol 166 (6) ◽  
pp. 1747-1757 ◽  
Author(s):  
S E Ratnofsky ◽  
A Peterson ◽  
J L Greenstein ◽  
S J Burakoff

In general, the human CD8 molecule is expressed on T cells specific for HLA class I molecules. Studies designed to delineate the function and to define the ligand of the CD8 molecule have been complicated by the fact that the presumptive ligand for CD8 is on the HLA class I molecule, the same molecule encoding the ligand for the antigen-specific T cell receptor. The ability to express genes in cells other than their natural host has produced a new technology with which to approach CD8 functional studies. The insertion of a cDNA clone for CD8 in a defective retroviral vector has allowed the transfer of CD8 by infection with the resulting defective retrovirus. CD8 was then expressed in an HLA class II-specific T cell, thus separating the ligand requirements of the TCR and CD8. By this approach, the human CD8 molecule was expressed in a murine T cell hybridoma specific for human class II antigens. The resulting CD8+ hybridomas demonstrated a 10-fold increase in IL-2 production over the parent cell line when stimulated with JY, a human B lymphoblastoid cell line expressing both class I and II HLA antigens, demonstrating that expression of CD8 increases T cell activation. mAbs directed against the CD8 molecule inhibited the response of CD8+ hybridomas to JY, supporting the conclusion that the CD8 molecule was fractional. The role of CD8 as a receptor for class I MHC antigens was addressed by stimulation with a cell line expressing HLA-DR antigens, but lacking the expression of HLA class I antigens (Daudi). Stimulation of the CD8+ hybridomas by Daudi did not result in increased IL-2 production. The response to Daudi was unaltered by the addition of anti-CD8 mAb, in contrast to the ability of anti-CD8 mAb to block JY stimulation. Furthermore, mAbs directed against the class I antigens present on JY cells were able to block the enhanced response of the CD8+ hybridomas to JY. These data support the hypothesis that HLA class I molecules are the ligands involved in the CD8-dependent enhancement of T cell activation.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4062-4062
Author(s):  
Daniel T Johnson ◽  
Ashley V Kroll ◽  
Ronnie H Fang ◽  
Justin Kline ◽  
Liangfang Zhang ◽  
...  

Abstract Acute Myeloid Leukemia (AML) is the most common acute leukemia in adults and has a five-year survival rate under 50%. Most patients will relapse even after complete remission is achieved through standard chemotherapy. Thus, one barrier in current AML therapy is how to target the minimal residual disease during remission. Recent developments in understanding cancer cell antigen presentation and immunosuppression have revealed the promise of cancer immunotherapy in activating immune responses to target residual disease. Each leukemia patient has a unique spectrum of cell surface antigens, which are mostly uncharacterized. If these antigens can be efficiently presented to the patient's immune system, immune responses to fight the leukemia can be significantly enhanced. We therefore sought to develop and characterize an AML cell membrane-coated nanoparticle (AMCNP) platform with nanoparticles (NPs) carrying the same surface antigens as the source leukemic cells for use as an anti-cancer vaccine. To demonstrate that our AMCNP vaccines enhance leukemia-specific antigen dendritic cell (DC) presentation and T-cell responses, we modified the C1498 murine AML cell line to express membrane-bound chicken ovalbumin (C1498-mOVA) as a model antigen. We confirmed that the C1498-mOVA line presents the OVA MHC class-I "SIINFEKL" antigen through flow-cytometry and LacZ B3Z T-cell activation assays. The C1498-mOVA line remained leukemogenic when injected into C57BL/6 mice, with survival times between 30 and 55 days. We generated both C1498 and C1498-mOVA membrane-coated nanoparticles, that were packaged with CpG oligo-deoxynucleotides (CpG) as an immune-stimulatory adjuvant. The final AMCNPs exhibit a core-shell structure with uniform coating as shown by transmission electron microscopy. The C1498-mOVA AMCNPs retained mOVA antigen. To confirm that the C1498-mOVA AMCNPs can effectively stimulate DC OVA MHC class I cross-presentation, we pulsed primary bone marrow derived DCs with C1498 AMCNPs or C1498-mOVA AMCNPs; only the C1498-mOVA AMCNP pulsed DCs specifically elicited OVA MHC class-I T-cell activation in lacZ B3Z T-cell activation assays. To verify that the AMCNPs can elicit antigen-specific immune responses in vivo, we vaccinated C57BL/6 mice with C1498 AMCNPs, C1498-mOVA AMCNPs, or equivalent amounts of whole cell lysates. When stimulated ex vivo with OVA peptide, immune-cell preparations from the C1498-mOVA AMCNP vaccinated mice showed significantly enhanced production of OVA-specific T-cells and IFN-γ, demonstrating increased immune responses. To assess if the enhanced cellular immunity afforded by the C1498-mOVA AMCNP formula can translate into functional rejection of leukemia cells, we performed a prophylactic study using the C1498-mOVA model. Mice vaccinated with the C1498-mOVA AMCNPs all survived beyond 120 days post C1498-mOVA cell challenge, compared to mock treated control mice which had a median survival of 60 days. Collectively, we developed an AMCNP platform that carries AML surface antigens and can be packaged with immunostimulatory adjuvants. These AMCNPs retained AML specific antigens, elicited enhanced antigen specific immune responses after in vivo vaccination, and improved immunity against AML challenge. Therefore, using AML cell membrane coated NPs to enhance anticancer immunity is a feasible strategy for AML vaccination immunotherapy. Disclosures Kline: iTeos: Research Funding; Merck: Honoraria, Research Funding.


1997 ◽  
Vol 62 (3) ◽  
pp. 287-291 ◽  
Author(s):  
Fabrizio Poccia ◽  
Miroslav Malkovsky ◽  
Marie Lise Gougeon ◽  
Marc Bonneville ◽  
Miguel Lopez-Botet ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document