HLA class I expression on the erythrocytes of cord and maternal blood — Flow cytometric analysis

1996 ◽  
Vol 47 (1-2) ◽  
pp. 110
Author(s):  
G. Paterakis ◽  
A. Germenis ◽  
D. Skoumi ◽  
N. Koutsodimas ◽  
C. Stavropoulos-Giokas
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 740-740
Author(s):  
Nanyan Zhang ◽  
Peter Newman

Abstract Antibodies that form against human platelet alloantigens (HPAs) are responsible for several clinically important alloimmune bleeding disorders, including fetal and neonatal alloimmune thrombocytopenia, posttransfusion purpura, and multitransfusion platelet refractoriness. Some of HPAs are relatively rare in the population, and difficult to obtain for purposes of transfusion therapy and diagnostic testing. In addition, HPA alloantisera often contain antibodies against human leucocyte antigen (HLA) class I, thereby limiting antibody detection to glycoprotein (GP)-specific assays such as the modified antigen capture enzyme-linked immunosorbent assay (MACE) and the monoclonal immobilization of platelet antigen (MAIPA), which are tedious and require solubilization of platelet GPs that may cause the loss of epitopes. In this study we aimed to generate gene-edited, HPA-specific, megakaryocytes (MKs) derived from human induced pluripotent stem cells (iPSCs) that could be used for simple flow cytometric detection of specific HPA alloantibodies present in patient sera. The HPA-3a/HPA-3b alloantigen system, also known as Baka/Bakb, is caused by a single T13809G nucleotide substitution in the ITGA2B gene, resulting in an Ile874Ser amino acid polymorphism near the C terminus of the integrin αIIb subunit (GPIIb). Here we targeted HPA-3 system because alloantibodies targeting HPA-3 are often hard to detect with current detection methods, in part due to the requirement for cell type-specific glycosylation. To prevent interference of anti-A or anti-B antibodies in patient sera, a blood type O iPSC line (OT1-1) was generated from human peripheral blood mononuclear cells derived from a healthy donor using integration-free episomal vectors. The gene for β2 microglobulin (B2M) was first ablated from the OT1-1 iPS cell line using CRISPR/Cas9 to prevent binding of HLA class I alloantibodies. The resulting B2M knockout (B2MKO) cells were then additionally gene edited to convert the endogenous HPA-3a alloantigenic epitope present on B2MKO OT1-1 cells to HPA-3b. Two different guide RNAs targeting sequences that flank exon 26 of the ITGA2B gene were designed such that the entire exon harboring the HPA-3 polymorphic site was removed. A plasmid harboring a template replacing exon 26 with the G13809 mutation, flanked by 600 bp homology arms, was cotransfected into the B2MKO OT1-1 iPSCs together with the two CRISPR/Cas9 guide RNA constructs. iPSC clones containing the desired targeted T13809G mutation were identified by a diagnostic MfeI digestion specific for the G13089-bearing HPA-3b allele. Sequence analysis confirmed conversion of T13089 to G in these HPA-3b clones. Flow cytometric analysis showed the HPA-3a iPSCs, when differentiated into CD41+/CD42b+ MKs, specifically reacted with HPA-3a, but not HPA-3b, patient sera, while the HPA-3b iPSC-derived MKs lost reactivity with HPA-3a patient serum, and gained the reactivity with HPA-3b patient sera. Taken together, we have established genetically modified iPSC-derived MKs expressing specific HPAs that are suitable for simple flow cytometry-based detection of HPA alloantibodies in patient sera, with low non-specific background binding. This system provides intact antigens on the cell surface with carbohydrate moieties that likely mimic those found on human platelets, thus facilitating the detection of HPA alloantibodies that are normally hard to detect with current methods. Application of this strategy to genetically edit this and other clinically-important HPAs holds great potential for producing Designer Platelets for diagnostic, investigative and ultimately therapeutic use. Disclosures No relevant conflicts of interest to declare.


PLoS ONE ◽  
2011 ◽  
Vol 6 (3) ◽  
pp. e17813 ◽  
Author(s):  
Urban Sester ◽  
Mathias Fousse ◽  
Jan Dirks ◽  
Ulrich Mack ◽  
Antje Prasse ◽  
...  

1988 ◽  
Vol 23 (2) ◽  
pp. 150 ◽  
Author(s):  
J. Szollosi ◽  
F.M. Brodsky ◽  
M. Balazs ◽  
P. Nagy ◽  
L. Tron ◽  
...  

Vox Sanguinis ◽  
1995 ◽  
Vol 68 (2) ◽  
pp. 109-120 ◽  
Author(s):  
J. Neumüller ◽  
M. Tohidast-Akrad ◽  
R. Jilch ◽  
D.W. M. Schwartz ◽  
W.R. Mayr

Blood ◽  
1999 ◽  
Vol 94 (9) ◽  
pp. 3094-3100 ◽  
Author(s):  
Kiyotaka Kuzushima ◽  
Yo Hoshino ◽  
Ken Fujii ◽  
Naoaki Yokoyama ◽  
Masatoshi Fujita ◽  
...  

Abstract We have developed an efficient and rapid method for detection of Epstein-Barr virus (EBV)-specific CD8+ T-cell frequencies both in freshly isolated peripheral blood mononuclear cells (PBMCs) and in vitro established cytotoxic T lymphocyte (CTL) lines. Responder cells are thereby stimulated with an autologous lymphoblastoid cell line for 5 hours and intracellular accumulation of interferon γ (IFNγ) is detected by multiparameter flow cytometric analysis. EBV-specific CD8+ T-cell frequencies ranged between 0.63% and 1.29% in PBMCs of 5 healthy long-term EBV carriers. Using EBV-specific T-cell lines, it was shown that flow cytometric analysis is more sensitive than limiting dilution analysis for CTL precursors and enzyme-linked immunospot assay detecting IFNγ-producing T cells. The class I restriction of IFNγ production was confirmed using an anti-class I monoclonal antibody (MoAb). Information on other cytokine production of EBV-specific CTLs could be obtained using combinations of anti-cytokine MoAbs. The sensitive and rapid nature of the flow cytometric assay for EBV-specific CD8+ T-cell frequency has significant advantages for evaluation of EBV-specific CD8+ T-cell responses in PBMCs of patients with EBV-related diseases.


2002 ◽  
Vol 70 (11) ◽  
pp. 6140-6146 ◽  
Author(s):  
Jürgen Rödel ◽  
Heinz Vogelsang ◽  
Katrin Prager ◽  
Matthias Hartmann ◽  
Karl-Hermann Schmidt ◽  
...  

ABSTRACT Chlamydia trachomatis infection can cause reactive arthritis that is associated with the persistence of chlamydial organisms in the joint. Fibroblasts of the synovial membrane represent host cells for Chlamydia during articular infection. In this study we investigated the expression of HLA class I molecules in synovial fibroblasts following infection with C. trachomatis D. The expression of HLA class I heavy chain (HLA-I) was up-regulated in infected cultures as shown by reverse transcription-PCR and immunoblotting. The increase in cell surface expression of HLA-I and β2 microglobulin on infected fibroblasts was demonstrated by flow cytometric analysis. Suppression of enhanced production of interferon-stimulated gene factor 3γ (ISGF3γ) in infected cell cultures by antisense oligonucleotide treatment reduced the level of HLA-I. Blocking antibodies to beta interferon (IFN-β) inhibited the Chlamydia-induced enhancement of both ISGF3γ and HLA-I. These findings show that the up-regulation of HLA-I in synovial fibroblasts infected with C. trachomatis is caused by the induction of IFN-β, which in turn stimulates the synthesis of ISGF3γ, a transcription factor participating in the regulation of the HLA-I gene. The IFN-β-mediated expression of HLA-I on Chlamydia-infected cells may be a regulatory factor in the immune response in chlamydial infections.


Cytometry ◽  
2000 ◽  
Vol 41 (4) ◽  
pp. 271-278 ◽  
Author(s):  
Susann Schweitzer ◽  
Angelika M. Schneiders ◽  
Bettina Langhans ◽  
Wolfgang Kraas ◽  
G�nther Jung ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (22) ◽  
pp. e1-e8 ◽  
Author(s):  
Nanyan Zhang ◽  
Sentot Santoso ◽  
Richard H. Aster ◽  
Brian R. Curtis ◽  
Peter J. Newman

Key Points HPA allele–specific HLA class I–negative MKs can be differentiated from CRISPR-edited human iPSCs. Such cells can be stored frozen and thawed to use in whole-cell flow cytometric assays to detect anti-HPA-3a, -3b, and -9b alloantibodies.


2019 ◽  
Vol 152 (Supplement_1) ◽  
pp. S151-S152
Author(s):  
Maryna Vazmitsel ◽  
Dong Chen ◽  
Barbara Gruner ◽  
Emily Coberly

Abstract Objectives Fetal/neonatal alloimmune thrombocytopenia (FNAIT) occurs when maternal IgG alloantibodies against paternal human platelet antigens (HPA) cross the placenta and cause the destruction of fetal platelets. The vast majority (up to 95%) of FNAIT cases are caused by antibodies against HPA-1a or HPA-5b antigens, while the remaining cases are usually due to antibodies against a variety of other HPA antigens. Cases of FNAIT due to anti-HLA antibodies are extremely uncommon and have only rarely been reported. We present a case of FNAIT suspected to be caused by anti-HLA class I alloantibodies. Methods The patient is a term infant boy born to a 32-year-old G2T2L2 mother. The mother had a previous diagnosis of Still disease (an adult form of systemic juvenile rheumatoid arthritis) but experienced complete resolution of symptoms and was off all treatment during the pregnancy. At birth, laboratory testing revealed isolated severe thrombocytopenia (platelet count 38,000/mcL) in an otherwise healthy-appearing infant. Results The infant had no evidence of bleeding, and testing for TORCH infection, sepsis, and DIC was negative. The maternal blood type was O positive. The maternal platelet count was normal. FNAIT was suspected and the infant was given two platelet transfusions from the same HPA 1a and 5b antigen-negative donor with no significant or sustained improvement in platelet count. Maternal platelet antibody testing subsequently revealed an absence of HPA antibodies, but anti-HLA class I alloantibodies were present. The infant was treated with three subsequent doses of IVIg with improvement in platelet count. No significant hemorrhage occurred. Conclusion HLA class I antibodies are commonly found in multiparous women but are not generally thought to cause significant fetal complications during subsequent pregnancies. This case suggests that, although rarely reported, HLA class I alloantibodies may be capable of causing FNAIT.


Sign in / Sign up

Export Citation Format

Share Document