Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(21)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126487

ABSTRACT

The human leukocyte antigen (HLA)-Ib molecule, HLA-F, is known as a CD4+ T-cell protein and mediator of HIV progression. While HLA-Ia molecules do not have the chance to select and present viral peptides for immune recognition due to protein downregulation, HLA-F is upregulated. Post HIV infection, HLA-F loses the affinity to its activating receptor KIR3DS1 on NK cells leading to progression of the HIV infection. Several studies aimed to solve the question of the biophysical interface between HLA ligands and their cognate receptors. It became clear that even an invariant HLA molecule can be structurally modified by the variability of the bound peptide. We recently discovered the ability of HLA-F to select and present peptides and the HLA-F allele-specific peptide selection from the proteomic content using soluble HLA (sHLA) technology and a sophisticated MS method. We established recombinant K562 cells that express membrane-bound HLA-F*01:01, 01:03 or 01:04 complexes. While a recombinant soluble form of KIR3DS1 did not bind to the peptide-HLA-F complexes, acid elution of the peptides resulted in the presentation of HLA-F open conformers, and the binding of the soluble KIR3DS1 receptor increased. We used CD4+/HIV- and CD4+/HIV+ cells and performed an MS proteome analysis. We could detect hemoglobin as significantly upregulated in CD4+ T-cells post HIV infection. The expression of cellular hemoglobin in nonerythroid cells has been described, yet HLA-Ib presentation of hemoglobin-derived peptides is novel. Peptide sequence analysis from HLA-F allelic variants featured hemoglobin peptides as dominant and shared. The reciprocal experiment of binding hemoglobin peptide fractions to the HLA-F open conformers resulted in significantly diminished receptor recognition. These results underpin the molecular involvement of HLA-F and its designated peptide ligand in HIV immune escape.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Hemoglobins/metabolism , Histocompatibility Antigens Class I/immunology , Peptide Fragments/metabolism , Proteome/analysis , Receptors, KIR3DS1/immunology , HIV Infections/metabolism , HIV Infections/virology , HIV-1/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Protein Binding , Receptors, KIR3DS1/metabolism
2.
Int J Mol Sci ; 21(12)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32575403

ABSTRACT

Natural killer (NK) cell therapies are a tool to antagonize a dysfunctional immune system. NK cells recognize malignant cells, traffic to a tumor location, and infiltrate the solid tumor. The immune checkpoint molecule human leukocyte antigen (HLA)-G is upregulated on malignant cells but not on healthy surrounding cells, the requirement of understanding the basis of receptor mediated events at the HLA-G/NK cell interface becomes obvious. The NK cell receptors ILT2 and KIR2DL4 have been described to bind to HLA-G; however, their differential function and expression levels on NK cell subsets suggest the existence of an unreported receptor. Here, we performed a ligand-based receptor capture on living cells utilizing sHLA-G*01:01 molecules coupled to TriCEPS and bound to NK cells followed by mass spectrometric analyses. We could define NKG2A/CD94 as a cognate receptor of HLA-G. To verify the results, we used the reciprocal method by expressing recombinant soluble heterodimeric NKG2A/CD94 molecules and used them to target HLA-G*01:01 expressing cells. NKG2A/CD94 could be confirmed as an immune receptor of HLA-G*01:01. Despite HLA-G is marginal polymorphic, we could previously demonstrate that the most common allelic subtypes HLA-G*01:01/01:03 and 01:04 differ in peptide repertoire, their engagement to NK cells, their catalyzation of dNK cell proliferation and their impact on NK cell development. Continuing these studies with regard to NKG2A/CD94 engagement we engineered recombinant single antigen presenting K562 cells and targeted the surface expressed HLA-G*01:01, 01:03 or 01:04 molecules with NKG2A/CD94. Specificity and sensitivity of HLA-G*01:04/NKG2A/CD94 engagement could be significantly verified. The binding affinity decreases when using K562-G*01:03 or K562-G*01:01 cells as targets. These results demonstrate that the ligand-receptor assignment between HLA-G and NKG2A/CD94 is dependent of the amino acid composition in the HLA-G heavy chain. Understanding the biophysical basis of receptor-mediated events that lead to NK cell inhibition would help to remove non-tumor reactive cells and support personalized mild autologous NK cell therapies.


Subject(s)
HLA-G Antigens/metabolism , Killer Cells, Natural/cytology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Amino Acid Motifs , Biotin/analogs & derivatives , Biotin/metabolism , Cell Line , Genetic Variation , HEK293 Cells , HLA-G Antigens/chemistry , HLA-G Antigens/genetics , Humans , Hydrazines/metabolism , K562 Cells , Killer Cells, Natural/immunology , Mass Spectrometry , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily D/genetics , Protein Binding , Succinimides/metabolism
3.
Int J Mol Sci ; 20(22)2019 Nov 08.
Article in English | MEDLINE | ID: mdl-31717259

ABSTRACT

Peptide-dependent engagement between human leucocyte antigens class I (HLA-I) molecules and their cognate receptors has been extensively analyzed. HLA-F belongs to the non-classical HLA-Ib molecules with marginal polymorphic nature and tissue restricted distribution. The three common allelic variants HLA-F*01:01/01:03/01:04 are distinguished by polymorphism outside the peptide binding pockets (residue 50, α1 or residue 251, α3) and are therefore not considered relevant for attention. However, peptide selection and presentation undergoes a most elaborated extraction from the whole available proteome. It is known that HLA-F confers a beneficial effect on disease outcome during HIV-1 infections. The interaction with the NK cell receptor initiates an antiviral downstream immune response and lead to delayed disease progression. During the time of HIV infection, HLA-F expression is upregulated, while its interaction with KIR3DS1 is diminished. The non-polymorphic nature of HLA-F facilitates the conclusion that understanding HLA-F peptide selection and presentation is essential to a comprehensive understanding of this dynamic immune response. Utilizing soluble HLA technology we recovered stable pHLA-F*01:01, 01:03 and 01:04 complexes from K562 cells and analyzed the peptides presented. Utilizing a sophisticated LC-MS-method, we analyzed the complete K562 proteome and matched the peptides presented by the respective HLA-F subtypes with detected proteins. All peptides featured a length of 8 to 24 amino acids and are not N-terminally anchored; the C-terminus is preferably anchored by Lys. To comprehend the alteration of the pHLA-F surface we structurally compared HLA-F variants bound to selected peptides. The peptides were selected from the same cellular content; however, no overlap between the proteomic source of F*01:01, 01:03 or 01:04 selected peptides could be observed. Recognizing the balance between HLA-F expression, HLA-F polymorphism and peptide selection will support to understand the role of HLA-F in viral pathogenesis.


Subject(s)
Alleles , Histocompatibility Antigens Class I/genetics , Peptides/metabolism , Proteomics , Amino Acid Motifs , Gene Ontology , Histocompatibility Antigens Class I/chemistry , Humans , K562 Cells , Protein Binding , Receptors, Natural Killer Cell/metabolism
4.
Pharmaceutics ; 11(10)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31618895

ABSTRACT

Drug hypersensitivity reactions that resemble acute immune reactions are linked to certain human leucocyte antigen (HLA) alleles. Severe and life-threatening Stevens Johnson Syndrome and Toxic Epidermal Necrolysis following treatment with the antiepileptic and psychotropic drug Carbamazepine are associated with HLA-B*15:02; whereas carriers of HLA-A*31:01 develop milder symptoms. It is not understood how these immunogenic differences emerge genotype-specific. For HLA-B*15:02 an altered peptide presentation has been described following exposure to the main metabolite of carbamazepine that is binding to certain amino acids in the F pocket of the HLA molecule. The difference in the molecular mechanism of these diseases has not been comprehensively analyzed, yet; and is addressed in this study. Soluble HLA-technology was utilized to examine peptide presentation of HLA-A*31:01 in presence and absence of carbamazepine and its main metabolite and to examine the mode of peptide loading. Proteome analysis of drug-treated and untreated cells was performed. Alterations in sA*31:01-presented peptides after treatment with carbamazepine revealed different half-life times of peptide-HLA- or peptide-drug-HLA complexes. Together with observed changes in the proteome elicited through carbamazepine or its metabolite these results illustrate the mechanistic differences in carbamazepine hypersensitivity for HLA-A*31:01 or B*15:02 patients and constitute the bridge between pharmacology and pharmacogenetics for personalized therapeutics.

5.
Int J Mol Sci ; 20(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344940

ABSTRACT

Human cytomegalovirus (HCMV) is ubiquitously prevalent. HCMV infection is typically asymptomatic and controlled by the immune system in healthy individuals, yet HCMV can be severely pathogenic for the fetus during pregnancy and in immunocompromised persons, such as transplant recipients or HIV infected patients. HCMV has co-evolved with the hosts, developed strategies to hide from immune effector cells and to successfully survive in the human organism. One strategy for evading or delaying the immune response is maintenance of the viral genome to establish the phase of latency. Furthermore, HCMV immune evasion involves the downregulation of human leukocyte antigens (HLA)-Ia molecules to hide infected cells from T-cell recognition. HCMV expresses several proteins that are described for downregulation of the HLA class I pathway via various mechanisms. Here, we review the wide range of immune evasion mechanisms of HCMV. Understanding the mechanisms of HCMV immune evasion will contribute to the development of new customized therapeutic strategies against the virus.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunity, Cellular/genetics , Killer Cells, Natural/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/virology , Genome, Viral/immunology , HLA Antigens/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Latency/immunology
6.
HLA ; 94(1): 25-38, 2019 07.
Article in English | MEDLINE | ID: mdl-30912293

ABSTRACT

T-cell receptors possess the unique ability to survey and respond to their permanently modified ligands, self HLA-I molecules bound to non-self peptides of various origin. This highly specific immune function is impaired following hematopoietic stem cell transplantation (HSCT) for a timespan of several months needed for the maturation of T-cells. Especially, the progression of HCMV disease in immunocompromised patients induces life-threatening situations. Therefore, the need for a new immune system that delivers vital and potent CD8+ T-cells carrying TCRs that recognize even one human cytomegalovirus (HCMV) peptide/HLA molecule and clear the viral infection long term becomes obvious. The transcription and translation of HCMV proteins in the lytic cycle is a precisely regulated cascade of processes, therefore, it is a highly sensitive challenge to adjust the exact time point of HCMV-peptide recruitment over self-peptides. We utilized soluble HLA technology in HCMV-infected fibroblasts and sequenced naturally sHLA-A*24:02 presented HCMV-derived peptides. One peptide of 14 AAs length derived from the IE2 antigen induced the strongest T-cell responses; this peptide can be detected with a low ranking score in general peptide prediction databanks. These results highlight the need for elaborate and HLA-allele specific peptide selection.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , HLA-A24 Antigen/immunology , Histocompatibility Antigens Class I/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Alleles , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Fibroblasts/immunology , Hematopoietic Stem Cell Transplantation , Humans
7.
Int J Mol Sci ; 20(6)2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30909402

ABSTRACT

On healthy cells the non-classical HLA class Ib molecule HLA-E displays the cognate ligand for the NK cell receptor NKG2A/CD94 when bound to HLA class I signal peptide sequences. In a pathogenic situation when HLA class I is absent, HLA-E is bound to a diverse set of peptides and enables the stimulatory NKG2C/CD94 receptor to bind. The activation of CD8⁺ T cells by certain p:HLA-E complexes illustrates the dual role of this low polymorphic HLA molecule in innate and adaptive immunity. Recent studies revealed a shift in the HLA-E peptide repertoire in cells with defects in the peptide loading complex machinery. We recently showed that HLA-E presents a highly diverse set of peptides in the absence of HLA class Ia and revealed a non-protective feature against NK cell cytotoxicity mediated by these peptides. In the present study we have evaluated the molecular basis for the impaired NK cell inhibition by these peptides and determined the cell surface stability of individual p:HLA-E complexes and their binding efficiency to soluble NKG2A/CD94 or NKG2C/CD94 receptors. Additionally, we analyzed the recognition of these p:HLA-E epitopes by CD8⁺ T cells. We show that non-canonical peptides provide stable cell surface expression of HLA-E, and these p:HLA-E complexes still bind to NKG2/CD94 receptors in a peptide-restricted fashion. Furthermore, individual p:HLA-E complexes elicit activation of CD8⁺ T cells with an effector memory phenotype. These novel HLA-E epitopes provide new implications for therapies targeting cells with abnormal HLA class I expression.


Subject(s)
Adaptive Immunity , Immunity, Innate , Immunomodulation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Histocompatibility Antigens Class I/immunology , Humans , NK Cell Lectin-Like Receptor Subfamily C/chemistry , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/chemistry , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Peptides/chemistry , Peptides/immunology , Protein Binding , Protein Multimerization , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , HLA-E Antigens
8.
Immunogenetics ; 68(4): 247-60, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26758079

ABSTRACT

Defining permissive and non-permissive mismatches for transplantation is a demanding challenge. Single mismatches at amino acid (AA) position 156 of human leucocyte antigen (HLA) class I have been described to alter the peptide motif, repertoire, or mode of peptide loading through differential interaction with the peptide-loading complex. Hence, a single mismatch can tip the balance and trigger an immunological reaction. HLA-B*35 subtypes have been described to evade the loading complex, 156 mismatch distinguishing B*35:01 and B*35:08 changes the binding groove sufficiently to alter the sequence features of the selected peptide repertoire. To understand the functional influences of residue 156 in B*35 variants, we analyzed the peptide binding profiles of HLA-B*35:01(156Leu), B*35:08(156Arg) and B*35:62(156Trp). The glycoprotein tapasin represents a target for immune evasions and functions within the multimeric peptide-loading complex to stabilize empty class I molecules and promote acquisition of high-affinity peptides. All three B*35 subtypes showed a tapasin-independent mode of peptide acquisition. HLA-B*35-restricted peptides of low- and high-binding affinities were recovered in the presence and absence of tapasin and subsequently sequenced utilizing mass spectrometry. The peptides derived from B*35 variants differ substantially in their features dependent on their mode of recruitment; all peptides were preferentially anchored by Pro at p2 and Tyr, Phe, Leu, or Lys at pΩ. However, the Trp at residue 156 altered the p2 motif to an Ala and restricted the pΩ to a Trp. Our results highlight the importance of understanding the impact of key micropolymorphism and how a single AA mismatch orchestrates the neighboring AAs.


Subject(s)
Amino Acid Substitution/genetics , HLA-B35 Antigen/genetics , Peptides/genetics , Polymorphism, Genetic/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Amino Acid Substitution/immunology , Amino Acids/genetics , Amino Acids/immunology , Cell Line , Endoplasmic Reticulum/immunology , HLA-B35 Antigen/immunology , Hematopoietic Stem Cell Transplantation , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Peptides/immunology , Protein Binding/immunology
9.
J Immunol Res ; 2014: 352160, 2014.
Article in English | MEDLINE | ID: mdl-25401109

ABSTRACT

The classical class I human leukocyte antigens (HLA-A, -B, and -C) present allele-specific self- or pathogenic peptides originated by intracellular processing to CD8(+) immune effector cells. Even a single mismatch in the heavy chain (hc) of an HLA class I molecule can impact on the peptide binding profile. Since HLA class I molecules are highly polymorphic and most of their polymorphisms affect the peptide binding region (PBR), it becomes obvious that systematic HLA matching is crucial in determining the outcome of transplantation. The opposite holds true for the nonclassical HLA class I molecule HLA-E. HLA-E polymorphism is restricted to two functional versions and is thought to present a limited set of highly conserved peptides derived from class I leader sequences. However, HLA-E appears to be a ligand for the innate and adaptive immune system, where the immunological response to peptide-HLA-E complexes is dictated through the sequence of the bound peptide. Structural investigations clearly demonstrate how subtle amino acid differences impact the strength and response of the cognate CD94/NKG2 or T cell receptor.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility/immunology , Protein Structure, Tertiary , Adaptive Immunity/immunology , Amino Acid Sequence , Histocompatibility Antigens Class I/metabolism , Humans , Immunity, Innate/immunology , Models, Molecular , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Protein Binding/immunology , HLA-E Antigens
10.
J Immunol Res ; 2014: 246171, 2014.
Article in English | MEDLINE | ID: mdl-25254222

ABSTRACT

HLA class I incompatibilities still remain one of the main barriers for unrelated bone marrow transplantation (BMT); hence the molecular understanding of how to mismatch patients and donors and still have successful clinical outcomes will guide towards the future of unrelated BMT. One way to estimate the magnitude of polymorphisms within the PBR is to determine which peptides can be selected by individual HLA alleles and subsequently presented for recognition by T cells. The features (structure, length, and sequence) of different peptides each confer an individual pHLA landscape and thus directly shape the individual immune response. The elution and sequencing of peptides by mass spectrometric analysis enable determining the bona fide repertoire of presented peptides for a given allele. This is an effective and simple way to compare the functions of allelic variants and make a first assessment of their degree of permissivity. We describe the methodology used for peptide sequencing and the limitations of peptide prediction tools compared to experimental methods. We highlight the altered peptide features that are observed between allelic variants and the need to discover the altered peptide repertoire in situations of "artificial" graft versus host disease (GvHD) that occur in HLA-specific hypersensitive immune responses to drugs.


Subject(s)
Bone Marrow Transplantation/methods , Graft vs Host Disease/immunology , HLA Antigens/immunology , Histocompatibility Testing/methods , Alleles , Graft vs Host Disease/genetics , HLA Antigens/genetics , Humans , Models, Immunological , Peptides/immunology , T-Lymphocytes/immunology , Unrelated Donors
11.
J Immunol Res ; 2014: 298145, 2014.
Article in English | MEDLINE | ID: mdl-25802875

ABSTRACT

While many HLA class I molecules interact directly with the peptide loading complex (PLC) for conventional loading of peptides certain class I molecules are able to present peptides in a way that circumvents the PLC components. We investigated micropolymorphisms at position 156 of HLA-A(*)24 allotypes and their effects on PLC dependence for assembly and peptide binding specificities. HLA-A(*)24:06(156Trp) and HLA-A(*)24:13(156Leu) showed high levels of cell surface expression while HLA-A(*)24:02(156Gln) was expressed at low levels in tapasin deficient cells. Peptides presented by these allelic variants showed distinct differences in features and repertoire. Immunoprecipitation experiments demonstrated all the HLA-A(*)24/156 variants to associate at similar levels with tapasin when present. Structurally, HLA-A(*)24:02 contains the residue triad Met97/His114/Gln156 and a Trp156 or Leu156 polymorphism provides tapasin independence by stabilizing these triad residues, thus generating an energetically stable and a more peptide receptive environment. Micropolymorphisms at position 156 can influence the generic peptide loading pathway for HLA-A(*)24 by altering their tapasin dependence for peptide selection. The trade-off for this tapasin independence could be the presentation of unusual ligands by these alleles, imposing significant risk following hematopoietic stem cell transplantation (HSCT).


Subject(s)
Amino Acids/genetics , Antigens/metabolism , HLA-A24 Antigen/metabolism , Peptide Fragments/metabolism , Antigen Presentation , Cell Line , Gene Knockdown Techniques , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Polymorphism, Genetic , Protein Binding , Protein Stability , Structure-Activity Relationship
12.
Immunogenetics ; 64(9): 663-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22706990

ABSTRACT

Knowledge about the magnitude of individual polymorphism is a critical part in understanding the complexity of comprehensive mismatching. HLA-B*44:09 differs from the highly frequent HLA-B*44:02 allele by amino acid exchanges at residues 77, 80, 81, 82 and 83. We aimed to identify the magnitude of these mismatches on the features of HLA-B*44:09 bound peptides since residues 77, 80 and 81 comprise part of the F pocket which determines sequence specificity at the pΩ position of the peptide. Using soluble HLA technology we determined >200 individual (nonduplicate) self-peptides from HLA-B*44:09 and compared their features with that of the published peptide features of HLA-B*44:02. Both alleles illustrate an anchor motif of E at p2. In contrast to the C-terminal peptide binding motif of B*44:02 (W, F, Y or L), B*44:09-derived peptides are restricted predominantly to L or F. The source of peptides for both alleles is identical (LCL 721.221 cells) allowing us to identify 23 shared peptides. The majority of these peptides however contained the restricted B*44:09 anchor motif of F or L at the pΩ position. Molecular modelling based on the B*44:02 structure highlights that the differences of the C-terminal peptide anchor between both alleles can be explained primarily by the B*44:02(81Ala) > B*44:09(81Leu) polymorphism which restricts the size of the amino acid that can be accommodated in the F pocket of B*44:09. These results highlight that every amino acid substitution has an impact of certain magnitude on the alleles function and demonstrate how surrounding residues orchestrate peptide specificity.


Subject(s)
Amino Acid Motifs , Amino Acids/metabolism , HLA-B44 Antigen/metabolism , Peptides/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/genetics , Binding Sites/genetics , HEK293 Cells , HLA-B44 Antigen/chemistry , HLA-B44 Antigen/genetics , Humans , Leucine/chemistry , Leucine/genetics , Leucine/metabolism , Models, Molecular , Peptides/chemistry , Peptides/genetics , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine/metabolism , Polymorphism, Genetic , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
13.
Immunogenetics ; 64(3): 245-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22009320

ABSTRACT

Position 45 represents a highly polymorphic residue within HLA class I alleles, which contacts the p2 position of bound peptides in 85% of the peptide-HLA structures analyzed, while the neighboring residues 41 and 46 are not involved in peptide binding. To investigate the influence of residue 45 at the functional level, we sequenced peptides eluted from recombinant HLA-B*44:08(41Ala/45Met/46Ala) molecules and compared their features with known peptides from B*44:02(41Thr/45Lys/46Glu). While HLA-B*44:02 has an anchor motif of E at the p2 anchor position, HLA-B*44:08 exhibits Q and L as anchor motif. The 45(Met/Lys) polymorphism contributes to the alteration in the peptide-binding motif and provides further evidence that mismatches at position 45 should be considered as nonpermissive in a transplantation setting.


Subject(s)
HLA-B Antigens/chemistry , HLA-B Antigens/genetics , Peptides/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution/immunology , HLA-B Antigens/metabolism , Histocompatibility/genetics , Histocompatibility/immunology , Humans , Models, Molecular , Peptides/metabolism , Protein Binding/immunology , Protein Conformation
14.
Haematologica ; 97(1): 98-106, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21993680

ABSTRACT

BACKGROUND: Polymorphic differences between donor and recipient human leukocyte antigen class I molecules can result in graft-versus-host disease due to distinct peptide presentation. As part of the peptide-loading complex, tapasin plays an important role in selecting peptides from the pool of potential ligands. Class I polymorphisms can significantly alter the tapasin-mediated interaction with the peptide-loading complex and although most class I allotypes are highly dependent upon tapasin, some are able to load peptides independently of tapasin. Several human leukocyte antigen B*44 allotypes differ exclusively at position 156 (B*44:02(156Asp), 44:03(156Leu), 44:28(156Arg), 44:35(156Glu)). From these alleles, only the high tapasin-dependency of human leukocyte antigen B*44:02 has been reported. DESIGN AND METHODS: We investigated the influence of position 156 polymorphisms on both the requirement of tapasin for efficient surface expression of each allotype and their peptide features. Genes encoding human leukocyte antigen B*44 variants bearing all possible substitutions at position 156 were lentivirally transduced into human leukocyte antigen class I-negative LCL 721.221 cells and the tapasin-deficient cell line LCL 721.220. RESULTS: Exclusively human leukocyte antigen B*44:28(156Arg) was expressed on the surface of tapasin-deficient cells, suggesting that the remaining B*44/156 variants are highly tapasin-dependent. Our computational analysis suggests that the tapasin-independence of human leukocyte antigen B*44:28(156Arg) is a result of stabilization of the peptide binding region and generation of a more peptide receptive state. Sequencing of peptides eluted from human leukocyte antigen B*44 molecules by liquid chromatography-electrospray ionization-mass spectrometry (LTQ-Orbitrap) demonstrated that both B*44:02 and B*44:28 share the same overall peptide motif and a certain percentage of their individual peptide repertoires in the presence and/or absence of tapasin. CONCLUSIONS: Here we report for the first time the influence of position 156 on the human leukocyte antigen/tapasin association. Additionally, the results of peptide sequencing suggest that tapasin chaperoning is needed to acquire peptides of unusual length.


Subject(s)
HLA-B44 Antigen/genetics , HLA-B44 Antigen/metabolism , Membrane Transport Proteins/metabolism , Peptides/metabolism , Alleles , Amino Acid Motifs/genetics , Amino Acid Sequence , Computational Biology/methods , Epitopes/chemistry , Epitopes/genetics , Gene Expression , Gene Silencing , HEK293 Cells , HLA-B44 Antigen/chemistry , Humans , Membrane Transport Proteins/genetics , Peptides/genetics , Polymorphism, Genetic , Protein Binding
15.
Hum Immunol ; 72(11): 1039-44, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21872626

ABSTRACT

Sequence variations outside exons 2 and 3 do not appear to affect the function of human leukocyte antigen (HLA) class I alleles. HLA-B*44:02:01:01 and -B*44:27 are considered functionally identical because they differ by a single amino acid substitution of Val > Ala at position 199, which is located in the α3 domain. To validate that HLA-B*44:02:01:01 and -B*44:27 represent functionally identical alleles that might reflect a permissive mismatch in hematopoetic stem cell transplantation (HSCT), we determined their peptide-binding features. B-lymphoblastic cells were lentivirally transduced with B*44:02 and B*44:27 constructs and soluble recombinant molecules were purified by affinity chromatography. Peptides were isolated and sequencing of single peptides was performed using liquid chromatography-electrospray ionization-tandem mass spectrometry (LTQ-Orbitrap) technology. We demonstrate that the peptide motif of B*44:02(199Val) and B*44:27(199Ala) is identical. Both variants feature E at P2 and Y, F, or W at PΩ in their ligands. Most of the identified peptides are 9 to 11 amino acids in length and approximately 20% of these ligands are shared between the alleles. Our results lead to the conclusion that B*44:02:01:01 and B*44:27 might have the same immune function, validating a theory that is now being used in deciding which donors to select in HSCT when there is no identical donor available.


Subject(s)
B-Lymphocytes/metabolism , HLA Antigens/metabolism , HLA-B44 Antigen/metabolism , Hematopoietic Stem Cell Transplantation , Peptide Fragments/metabolism , Amino Acid Motifs/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Line, Tumor , Chromatography, Affinity , Donor Selection , Exons/genetics , HLA Antigens/chemistry , HLA Antigens/genetics , HLA-B Antigens/genetics , HLA-B Antigens/metabolism , HLA-B44 Antigen/chemistry , HLA-B44 Antigen/genetics , Histocompatibility/immunology , Humans , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Polymorphism, Genetic , Protein Binding/genetics , Protein Binding/immunology , Tandem Mass Spectrometry , Transgenes/genetics
16.
Transfusion ; 47(12): 2330-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17764507

ABSTRACT

BACKGROUND: Binding of CCAAT-binding factor NF-Y (CBF/NF-Y) to a 43-bp repeat unit in the minisatellite region in the 5' region of the ABO gene (CBF/NF-Y enhancer region) plays an important role in regulating the transcription of ABO genes. The common ABO alleles were found to have CBF/NF-Y enhancer regions with specific numbers of 43-bp minisatellite repeats. MATERIAL AND METHODS: Blood samples from four healthy blood donors with weak B phenotypes were subjected to extensive ABO genotyping, including nucleotide sequencing of the 5' regulatory region containing the CBF/NF-Y enhancer. RESULTS: The coding region of the ABO genes exhibited common ABO*B101-heterozygous genotypes in all samples, but unexpected variations were observed in the CBF/NF-Y enhancer region. In two cases, the CBF/NF-Y enhancer motifs did not exhibit the expected ABO allele dependency. One, an AB(weak) sample was heterozygous for ABO*A101 and ABO*B101 but homozygous for the ABO*B101-specific CBF/NF-Y motif. The second had a common ABO*B101/ABO*O01 genotype but was heterozygous for ABO*A101- and ABO*O01-specific enhancer motifs. In the other two samples, novel CBF/NF-Y motifs were found. One contained a shortened version of an otherwise ABO*B101-specific CBF/NF-Y motif, and the other had a single-base substitution located 12 bp upstream from the beginning of the first 43-bp repeat of an ABO*B101-specific CBF/NF-Y enhancer sequence. CONCLUSION: The frequency of variations in the CBF/NF-Y region of the ABO gene in these samples with presumably common ABO*B101 alleles suggests that weak blood group B phenotypes may be caused by sequence variations in the CBF/NF-Y regulatory region.


Subject(s)
ABO Blood-Group System/genetics , CCAAT-Binding Factor/metabolism , Enhancer Elements, Genetic/genetics , Alleles , Genotype , Humans , Mutation , Phenotype , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics
17.
Immunogenetics ; 59(4): 253-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17294180

ABSTRACT

There have been several attempts over the years to identify positions in the peptide-binding region (PBR) of human leukocyte antigens (HLA) that influence the specificity of bound amino acids (AAs) at each position in the peptide. Originally, six pockets (A-F) were defined by calculating the surface area of the PBR on the crystal structure of HLA-A2 molecules. More recent crystallographic analyses of a variety of HLA alleles have led to broader pocket definitions. In this study, we examined the peptide-binding specificity of HLA-B*41 alleles and compared our results with the available pocket definitions. By generating recombinant HLA-B molecules and studying the eluted peptides by mass spectrometry and pool sequencing, we detected two different POmega peptide motifs within the B*41 group: Leu vs Val/Pro. Specificity was dependent on the presence of Leu (B*4102, B*4103, and B*4104) vs Trp (B*4101, B*4105, and B*4106) at AA position 95 in the HLA molecule, whose impact on POmega has been a subject of controversy in current pocket definitions. In contrast, the Arg97Ser mutation did not affect pocket F binding specificity in B*41 subtypes although residue 97 was previously identified as a modulator of peptide binding for several HLA class I alleles. According to most pocket definitions, this study shows that the Asn80Lys substitution in B*4105 impels the peptide's POmega anchor toward more promiscuity. Our sequencing results of peptides eluted from HLA-B*41 variants demonstrate the limitations of current pocket definitions and underline the need for an extended peptide motif database for improved understanding of peptide-major histocompatibility complex interactions.


Subject(s)
Amino Acids/chemistry , HLA-B Antigens/chemistry , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acids/genetics , HLA-B Antigens/genetics , Humans , Peptides/chemistry , Peptides/genetics , Protein Conformation
18.
Transfusion ; 47(1): 133-46, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17207242

ABSTRACT

BACKGROUND: Semaphorin 7A (Sema7A), the protein that carries the JMH blood group antigen, is involved in immune responses and plays an important role in axon growth and guidance. Because previous serologic studies on red blood cells (RBCs) suggested a considerable diversity of Sema7A, the present study was designed to elucidate the complex picture of the molecular diversity of this protein. STUDY DESIGN AND METHODS: The JMH antigen status was determined by serology, flow cytometry, and Western blot. Genomic and transcript analysis of SEMA7A was performed by nucleotide sequencing. Recombinant Sema7A proteins were used for genotype-phenotype correlation. A three-dimensional model of Sema7A was generated for topologic analyses. RESULTS: Our studies on 44 individuals with unusual JMH phenotypes and their family members revealed that aberrant Sema7A expression can be an inherited or an acquired phenomenon and is based on reduced surface expression or qualitative changes in Sema7A. These different phenotypes are caused by variations of the SEMA7A gene or seem to be generated by autoimmune-related or RBC lineage-specific mechanisms. The variant JMH phenotypes were related to the presence of missense mutations in SEMA7A, predicting amino acid changes in the semaphorin domain of Sema7A. Sequence analysis of the variant SEMA7A alleles revealed mutations affecting codons 207 and 460/461. Topologic analyses showed that Sema7A polymorphisms were prominently located on the top and bottom of the semaphorin domain, suggesting a functional relevance of these sites. CONCLUSION: These findings provide a basis with which to delineate the various ligand-binding surfaces of Sema7A.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/genetics , Semaphorins/chemistry , Semaphorins/genetics , Antigens, CD/blood , Cell Line, Transformed , Erythrocytes/metabolism , Flow Cytometry , GPI-Linked Proteins , Genetic Variation , Humans , Models, Molecular , Molecular Conformation , Mutation, Missense , Pedigree , Phenotype , Polymorphism, Genetic , Semaphorins/blood
19.
Transfusion ; 46(3): 434-40, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16533287

ABSTRACT

BACKGROUND: Weak blood group A and B phenotypes are correlated with ABO glycosyltransferases exhibiting single-amino-acid changes and/or C-terminal modifications. STUDY DESIGN AND METHODS: A healthy donor diagnosed as having weak A antigen expression and his two children were subjected to extensive ABO typing. HeLa cells were used to transfect ABO expression plasmids. RESULTS: The donor's red blood cells were type A(weak)B and his serum sample contained weakly reactive anti-A(1) antibodies. A single T>C transition identified at the +2 position of the start codon of an ABO*A101 allele predicted the disruption of this methionine codon. In the transfection studies, a significant reduction of A activity was observed on HeLa cells transfected with a plasmid containing the variant ABO*A allele. Coexpression of the respective antithetical ABO*B101 wild-type construct further reduced cell surface A antigen expression. Similar expression results were obtained with ABO constructs in which the Met(1) start codon and five alternative start sites at codons 20, 26, 43, 53, and 69 had successively been interrupted. CONCLUSION: The donor's weak blood group A phenotype most likely resulted from expression of an N-truncated A transferase triggered by alternative translation start sites in the transmembrane domain or stem region.


Subject(s)
ABO Blood-Group System/genetics , Alleles , Codon, Initiator/genetics , Glycosyltransferases/genetics , Point Mutation , Protein Biosynthesis/genetics , Blood Grouping and Crossmatching/methods , Female , Gene Expression , HeLa Cells , Humans , Male , Pedigree , Phenotype , Plasmids , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary , Transfection/methods
20.
Immunogenetics ; 56(10): 769-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15592665

ABSTRACT

The peptide motif of HLA-A*6603 was determined and compared with the available data on the peptide motifs of A*6601 and A*6602. A*6601 differs from A*6602 by two amino acids at positions 90 (Asp90Ala; outer loop) and 163 (Arg163Glu; pocket A). A*6603 differs from A*6601 and A*6602 by a single amino-acid exchange at position 70 (His70Gln; pockets A, B and C). No significant differences were found between the A*6602 and A*6603 peptide motifs suggesting that the Gln70His variation is of minor importance. However, the auxiliary anchors at position P1 of peptides bound by A*6601 (polar/acidic: Asp, Glu) and A*6602/6603 (polar/neutral: Ser) had striking differences. This finding may be best explained by the Arg163Glu substitution that results in a shift towards higher acidity in pocket A of A*6602/6603, apparently leading to the loss of preference for acidic auxiliary anchors. The similarity of A*6602 and A*6603 peptide motifs suggests low allogenicity when mismatched in stem cell transplantation. Inversely, the differences in A*6601 versus A*6602/6603 peptide motifs suggest that mismatches will have a higher allogenicity. These data will contribute to both assessing permissive mismatches in the A*66 group and weighting the impact of this individual amino-acid variation for matching and peptide binding algorithms.


Subject(s)
HLA-A Antigens/chemistry , Amino Acid Motifs , Binding Sites , HLA-A Antigens/metabolism , Humans , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...