CLL Cell Proliferation and IL-6 Production Are Regulated by CD160 - MHC Class I Interactions Offering New Therapeutic Targets.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1126-1126
Author(s):  
Jerome Giustiniani ◽  
Catherine Wiseman ◽  
Timothy Farren ◽  
John G. Gribben ◽  
Samir G. Agrawal

Abstract We have previously demonstrated the expression of the Natural Killer cell (NK) marker CD160 in CLL, which is not expressed by normal B cells1. CD160 is a cell surface molecule expressed by human and mouse circulating cytotoxic lymphocytes and exhibits a broad specificity for major histocompatibility complex (MHC) class Ia and Ib molecules 2,3,4. Triggering of CD160 on NK cells leads to cell proliferation and IL-6 production5. IL-6 is a pleiotropic cytokine produced by a variety of cell types, including lymphocytes6. IL-6 expression has been associated with the development of lymphomas7, while higher serum IL-6 levels correlated with shorter survival in CLL8. Without stimulation, CLL cells secreted low basal levels of INF-γ (100pg/ml) and even lower levels of IL-2, IL-4, IL-6, IL-10 and TNF-α (< 50pg/ml) (cytokine bead array, BD Bioscience). Incubation with CL1-R2, an anti-CD160 monoclonal antibody, led to an increase in IL-6 alone (up to 1500pg/ml), which was associated with significant cell proliferation (detected by 3H incorporation) - median 100%, range 50 to 400% (n = 13). Different patterns of response were seen, with all cases showing proliferation to CL1-R2 alone and with the positive control pan anti-immunoglobulin Ab (anti-Ig Ab). In some cases, there was marked synergy between CL1-R2 and anti-Ig Ab. We found that the MEC I and MEC II B-cell lines, derived from a patient with prolymphocytic progression of CLL (DSMZ Institut), express CD160 and show CL1-R2 enhanced proliferation (30 to 55% increase). However, MEC I and MEC II only unregulated IL-10 production (13 to 33% increase). Interestingly, proliferation of CD160Neg Sanchez EBV-immortalized B-cells was inhibited when these cells were incubated with CHO-CD160 transfectants. This inhibition was reversed with an anti-CD160 Ab or anti-MHC-class I W6/32 Ab. The CD160-MHC-I interaction plays a role in CLL biology. MAb engagement of CD160 leads to: a direct proliferative signal; CD160-triggered production of IL-6 and IL-6 mediated effects; inhibition of a constitutive negative signal mediated via MHC-I molecules by their ligand CD160 (supported by the inhibition of the Sanchez cells by CD160 interaction with their MHC class I). The CD160-MHC class I axis represents a new pathway in CLL biology offering new therapeutic targets. This work also suggests a role for targeting the IL-6/IL-6R system in CLL - for example, with humanized anti-IL6 R (Actemra), already used for Castleman’s disease and Rheumatoid Arthritis. Fig 1: cells were incubated 72h in medium only or completed with IgG (control), anti-CD 160 antibody (CL1-R2), anti-IgM, G, A antibody (PAN antibody) or mix with PAN antibody and CL1-R2. For cytokine expriments, supernatants were taken after 24 hours. (left scale: CPM value, right scale: IL-6 concentration in pg/ml) Fig 1:. cells were incubated 72h in medium only or completed with IgG (control), anti-CD 160 antibody (CL1-R2), anti-IgM, G, A antibody (PAN antibody) or mix with PAN antibody and CL1-R2. For cytokine expriments, supernatants were taken after 24 hours. (left scale: CPM value, right scale: IL-6 concentration in pg/ml)

1999 ◽  
Vol 189 (3) ◽  
pp. 493-500 ◽  
Author(s):  
Mary C. Nakamura ◽  
Paul A. Linnemeyer ◽  
Eréne C. Niemi ◽  
Llewellyn H. Mason ◽  
John R. Ortaldo ◽  
...  

Although activation of natural killer (NK) cytotoxicity is generally inhibited by target major histocompatibility complex (MHC) class I expression, subtle features of NK allorecognition suggest that NK cells possess receptors that are activated by target MHC I. The mouse Ly-49D receptor has been shown to activate NK cytotoxicity, although recognition of MHC class I has not been demonstrated previously. To define Ly-49D–ligand interactions, we transfected the mouse Ly-49D receptor into the rat NK line, RNK-16 (RNK.mLy-49D). As expected, anti– Ly-49D monoclonal antibody 12A8 specifically stimulated redirected lysis of the Fc receptor– bearing rat target YB2/0 by RNK.mLy-49D transfectants. RNK.mLy-49D effectors were tested against YB2/0 targets transfected with the mouse MHC I alleles H-2Dd, Db, Kk, or Kb. RNK.mLy-49D cells lysed YB2/0.Dd targets more efficiently than untransfected YB2/0 or YB2/0 transfected with Db, Kk, or Kb. This augmented lysis of H-2Dd targets was specifically inhibited by F(ab′)2 anti–Ly-49D (12A8) and F(ab′)2 anti–H-2Dd (34-5-8S). RNK.mLy-49D effectors were also able to specifically lyse Concanavalin A blasts isolated from H-2d mice (BALB/c, B10.D2, and DBA/2) but not from H-2b or H-2k mice. These experiments show that the activating receptor Ly-49D specifically interacts with the MHC I antigen, H-2Dd, demonstrating the existence of alloactivating receptors on murine NK cells.


2020 ◽  
Author(s):  
Xizheng Sun ◽  
Reika Tokunaga ◽  
Yoko Nagai ◽  
Ryo Miyahara ◽  
Akihiro Kishimura ◽  
...  

<p><a></a><a></a><a>We have validated that ligand peptides designed from antigen peptides could be used for targeting specific major histocompatibility complex class I (MHC-I)</a> molecules on cell surface. To design the ligand peptides, we used reported antigen peptides for each MHC-I molecule with high binding affinity. From the crystal structure of the peptide/MHC-I complexes, we determined a modifiable residue in the antigen peptides and replaced this residue with a lysine with an ε-amine group modified with functional molecules. The designed ligand peptides successfully bound to cells expressing the corresponding MHC-I molecules via exchange of peptides bound to the MHC-I. We demonstrated that the peptide ligands could be used to transport a protein or a liposome to cells expressing the corresponding MHC-I. The present strategy may be useful for targeted delivery to cells overexpressing MHC-I, which have been observed autoimmune diseases.</p>


2021 ◽  
Vol 12 ◽  
Author(s):  
Silvia D’Amico ◽  
Valerio D’Alicandro ◽  
Mirco Compagnone ◽  
Patrizia Tempora ◽  
Giusy Guida ◽  
...  

The endoplasmic reticulum aminopeptidase ERAP1 regulates innate and adaptive immune responses by trimming peptides for presentation by major histocompatibility complex (MHC) class I molecules. Previously, we have shown that genetic or pharmacological inhibition of ERAP1 on murine and human tumor cell lines perturbs the engagement of NK cell inhibitory receptors Ly49C/I and Killer-cell Immunoglobulin-like receptors (KIRs), respectively, by their specific ligands (MHC class I molecules), thus leading to NK cell killing. However, the effect of ERAP1 inhibition in tumor cells was highly variable, suggesting that its efficacy may depend on several factors, including MHC class I typing. To identify MHC class I alleles and KIRs that are more sensitive to ERAP1 depletion, we stably silenced ERAP1 expression in human HLA class I-negative B lymphoblastoid cell line 721.221 (referred to as 221) transfected with a panel of KIR ligands (i.e. HLA-B*51:01, -Cw3, -Cw4 and -Cw7), or HLA-A2 which does not bind any KIR, and tested their ability to induce NK cell degranulation and cytotoxicity. No change in HLA class I surface expression was detected in all 221 transfectant cells after ERAP1 depletion. In contrast, CD107a expression levels were significantly increased on NK cells stimulated with 221-B*51:01 cells lacking ERAP1, particularly in the KIR3DL1-positive NK cell subset. Consistently, genetic or pharmacological inhibition of ERAP1 impaired the recognition of HLA-B*51:01 by the YTS NK cell overexpressing KIR3DL1*001, suggesting that ERAP1 inhibition renders HLA-B*51:01 molecules less eligible for binding to KIR3DL1. Overall, these results identify HLA-B*51:01/KIR3DL1 as one of the most susceptible combinations for ERAP1 inhibition, suggesting that individuals carrying HLA-B*51:01-like antigens may be candidates for immunotherapy based on pharmacological inhibition of ERAP1.


1994 ◽  
Vol 3 (4) ◽  
pp. 297-307 ◽  
Author(s):  
Jacques Robert ◽  
Chantal Guiet ◽  
Louis Du Pasquier

Three new lymphoid tumors offering an assortment of variants in terms of MHC class I expressions, MHC class II expression, and Ig gene transcription have been discovered in the amphibianXenopus. One was developed in an individual of the isogenic LG15 clone (LG15/0), one in a frog of the LG15/40 clone (derived from a small egg recombinant of LG15), and one (ff-2) in a maleffsib of the individual in which MAR1, the first lymphoid tumor in Xenopus was found 2 years ago. These tumors developed primarily as thymus outgrowths and were transplantable in histocompatible tadpoles but not in nonhistocompatible hosts. Whereas LG15/0 and LG15/40 tumor cells also grow in adult LG15 frogs, theff-2 tumor, like the MAR1 cell line, is rejected by adultffanimals. Using flow cytometry with fluorescence-labeled antibodies and immunoprecipitation analysis, we could demonstrate that, like MAR1, these three new tumors express on their cell surface lymphopoietic markers recognized by mAbs FIF6 and RC47, as well as T-cell lineage markers recognized by mAbs AM22 (CD8-1ike) and X21.2, but not by immunologobulin (Ig) nor MHC class II molecules. Another lymphocyte-specific marker AM15 is expressed by 15/0 and 15/40 but notff-2 tumor cells. Theff-2 tumor cell expresses MHC class molecule in association withβ2-microglobulin on the surface, 15/40 cells contain cytoplasmic Iαchain that is barely detected at the cell surface by fluocytometry, and 15/0 cells do not synthesize class Iαchain at all. The three new tumors all produce large amounts of IgM mRNA of two different sizes but no Ig protein on the membrane nor in the cytoplasm. All tumor cell types synthesize large amount of Myc mRNA and MHC class I-like transcripts considered to be non classical.


1995 ◽  
Vol 60 (3) ◽  
pp. 281-286 ◽  
Author(s):  
MARGARITA SALCEDO ◽  
FETTER HOGLUND ◽  
HANS-GUSTAF LJUNGGREN

2019 ◽  
Vol 2019 ◽  
pp. 1-9 ◽  
Author(s):  
Simo Xia ◽  
Yijie Tao ◽  
Likun Cui ◽  
Yizhi Yu ◽  
Sheng Xu

MHC class I molecules are key in the presentation of antigen and initiation of adaptive CD8+ T cell responses. In addition to its classical activity, MHC I may possess nonclassical functions. We have previously identified a regulatory role of MHC I in TLR signaling and antibacterial immunity. However, its role in innate antiviral immunity remains unknown. In this study, we found a reduced viral load in MHC I-deficient macrophages that was independent of type I IFN production. Mechanically, MHC I mediated viral suppression by inhibiting the type I IFN signaling pathway, which depends on SHP2. Cross-linking MHC I at the membrane increased SHP2 activation and further suppressed STAT1 phosphorylation. Therefore, our data revealed an inhibitory role of MHC I in type I IFN response to viral infection and expanded our understanding of MHC I and antigen presentation.


Sign in / Sign up

Export Citation Format

Share Document