Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
HLA ; 102(4): 436-448, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37370222

RESUMO

HLA-sensitized patients on the transplant waiting list harbor antibodies and memory B cells directed against allogeneic HLA molecules, which decreases the chance to receive a compatible donor organ. Current desensitization strategies non-specifically target circulating antibodies and B cells, warranting the development of therapies that specifically affect HLA-directed humoral immune responses. We developed Chimeric HLA Antibody Receptor (CHAR) constructs comprising the extracellular part of HLA-A2 or HLA-A3 coupled to CD28-CD3ζ domains. CHAR-transduced cells expressing reporter constructs encoding T-cell activation markers, and CHAR-transduced CD8+ T cells from healthy donors were stimulated with HLA-specific monoclonal antibody-coated microbeads, and HLA-specific B cell hybridomas. CHAR T cell activation was measured by upregulation of T cell activation markers and IFNγ secretion, whereas CHAR T cell killing of B cell hybridomas was assessed in chromium release assays and by IgG ELISpot. HLA-A2- and HLA-A3-CHAR expressing cells were specifically activated by HLA-A2- and HLA-A3-specific monoclonal antibodies, either soluble or coated on microbeads, as shown by CHAR-induced transcription factors. HLA-A2 and HLA-A3 CHAR T cells efficiently produced IFNγ with exquisite specificity and were capable of specifically lysing hybridoma cells expressing HLA-A2- or HLA-A3-specific B-cell receptors, respectively. Finally, we mutated the α3 domain of the CHAR molecules to minimize any alloreactive T-cell reactivity against CHAR T cells, while retaining CHAR activity. These data show proof of principle for CHAR T cells to serve as precision immunotherapy to specifically desensitize (highly) sensitized solid organ transplant candidates and to treat antibody-mediated rejection after solid organ transplantation.


Assuntos
Anticorpos , Linfócitos B , Dessensibilização Imunológica , Transplante de Rim , Anticorpos/genética , Anticorpos/imunologia , Aloenxertos/imunologia , Linfócitos T , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/metabolismo , Interferon gama/imunologia , Citotoxicidade Imunológica , Linfócitos B/imunologia , Dessensibilização Imunológica/métodos , Estudo de Prova de Conceito , Linhagem Celular , Doadores de Sangue , Humanos
2.
J Virol ; 94(9)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32075930

RESUMO

The major histocompatibility complex (MHC) class I region of humans, chimpanzees (Pan troglodytes), and bonobos (Pan paniscus) is highly similar, and orthologues of HLA-A, -B, and -C are present in both Pan species. Based on functional characteristics, the different HLA-A allotypes are classified into different supertypes. One of them, the HLA A03 supertype, is widely distributed among different human populations. All contemporary known chimpanzee and bonobo MHC class I A allotypes cluster genetically into one of the six HLA-A families, the HLA-A1/A3/A11/A30 family. We report here that the peptide-binding motif of the Patr-A*05:01 allotype, which is commonly present in a cohort of western African chimpanzees, has a strong preference for binding peptides with basic amino acids at the carboxyl terminus. This phenomenon is shared with the family members of the HLA A03 supertype. Based on the chemical similarities in the peptide-binding pocket, we inferred that the preference for binding peptides with basic amino acids at the carboxyl terminus is widely present among the human, chimpanzee, and bonobo MHC-A allotypes. Subsequent in silico peptide-binding predictions illustrated that these allotypes have the capacity to target conserved parts of the proteome of human immunodeficiency virus type 1 (HIV-1) and the simian immunodeficiency virus SIVcpz.IMPORTANCE Most experimentally infected chimpanzees seem to control an HIV-1 infection and are therefore considered to be relatively resistant to developing AIDS. Contemporary free-ranging chimpanzees may carry SIVcpz, and there is evidence for AIDS-like symptoms in these free-ranging animals, whereas SIV infections in bonobos appear to be absent. In humans, the natural control of an HIV-1 infection is strongly associated with the presence of particular HLA class I allotypes. The ancestor of the contemporary living chimpanzees and bonobos survived a selective sweep targeting the MHC class I repertoire. We have put forward a hypothesis that this may have been caused by an ancestral retroviral infection similar to SIVcpz. Characterization of the relevant MHC allotypes may contribute to understanding the shaping of their immune repertoire. The abundant presence of MHC-A allotypes that prefer peptides with basic amino acids at the C termini suggests that these molecules may contribute to the control of retroviral infections in humans, chimpanzees, and bonobos.


Assuntos
Genes MHC Classe I/imunologia , Antígeno HLA-A3/imunologia , Primatas/imunologia , Alelos , Sequência de Aminoácidos , Animais , HIV-1/imunologia , Antígeno HLA-A3/metabolismo , Antígenos de Histocompatibilidade , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Pan paniscus/imunologia , Pan troglodytes/imunologia , Peptídeos/metabolismo , Filogenia , Ligação Proteica/imunologia , Infecções por Retroviridae/imunologia , Vírus da Imunodeficiência Símia/imunologia
3.
BMC Genomics ; 19(Suppl 1): 42, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29363421

RESUMO

BACKGROUND: Ebolavirus (EBOV) is responsible for one of the most fatal diseases encountered by mankind. Cellular T-cell responses have been implicated to be important in providing protection against the virus. Antigenic variation can result in viral escape from immune recognition. Mapping targets of immune responses among the sequence of viral proteins is, thus, an important first step towards understanding the immune responses to viral variants and can aid in the identification of vaccine targets. Herein, we performed a large-scale, proteome-wide mapping and diversity analyses of putative HLA supertype-restricted T-cell epitopes of Zaire ebolavirus (ZEBOV), the most pathogenic species among the EBOV family. METHODS: All publicly available ZEBOV sequences (14,098) for each of the nine viral proteins were retrieved, removed of irrelevant and duplicate sequences, and aligned. The overall proteome diversity of the non-redundant sequences was studied by use of Shannon's entropy. The sequences were predicted, by use of the NetCTLpan server, for HLA-A2, -A3, and -B7 supertype-restricted epitopes, which are relevant to African and other ethnicities and provide for large (~86%) population coverage. The predicted epitopes were mapped to the alignment of each protein for analyses of antigenic sequence diversity and relevance to structure and function. The putative epitopes were validated by comparison with experimentally confirmed epitopes. RESULTS & DISCUSSION: ZEBOV proteome was generally conserved, with an average entropy of 0.16. The 185 HLA supertype-restricted T-cell epitopes predicted (82 (A2), 37 (A3) and 66 (B7)) mapped to 125 alignment positions and covered ~24% of the proteome length. Many of the epitopes showed a propensity to co-localize at select positions of the alignment. Thirty (30) of the mapped positions were completely conserved and may be attractive for vaccine design. The remaining (95) positions had one or more epitopes, with or without non-epitope variants. A significant number (24) of the putative epitopes matched reported experimentally validated HLA ligands/T-cell epitopes of A2, A3 and/or B7 supertype representative allele restrictions. The epitopes generally corresponded to functional motifs/domains and there was no correlation to localization on the protein 3D structure. These data and the epitope map provide important insights into the interaction between EBOV and the host immune system.


Assuntos
Ebolavirus/imunologia , Mapeamento de Epitopos/métodos , Epitopos de Linfócito T/imunologia , Doença pelo Vírus Ebola/imunologia , Proteoma/imunologia , Proteínas Virais/imunologia , Ebolavirus/isolamento & purificação , Variação Genética , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/genética , Antígeno HLA-A3/imunologia , Antígeno HLA-A3/metabolismo , Antígeno HLA-B7/genética , Antígeno HLA-B7/imunologia , Antígeno HLA-B7/metabolismo , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Proteoma/metabolismo , Linfócitos T Citotóxicos/imunologia
4.
Cancer Immunol Immunother ; 65(11): 1339-1352, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27576783

RESUMO

Survivin is an anti-apoptotic protein that is highly expressed in many cancers, including malignant gliomas. Preclinical studies established that the conjugated survivin peptide mimic SurVaxM (SVN53-67/M57-KLH) could stimulate an anti-tumor immune response against murine glioma in vivo, as well as human glioma cells ex vivo. The current clinical study was conducted to test safety, immunogenicity and clinical effects of the vaccine. Recurrent malignant glioma patients whose tumors were survivin-positive, and who had either HLA-A*02 or HLA-A*03 MHC class I allele-positivity, were given subcutaneous injections of SurVaxM (500 µg) in Montanide ISA 51 with sargramostim (100 µg) at 2-week intervals. SurVaxM was well tolerated with mostly grade one adverse events (AE) and no serious adverse events (SAE) attributable to the study drug. Six patients experienced local injection site reactions; three patients reported fatigue (grades 1 and 2), and 2 patients experienced myalgia (grade 1). Six of eight immunologically evaluable patients developed both cellular and humoral immune responses to vaccine. The vaccine also stimulated HLA-A*02, HLA-A*03 and HLA-A*24 restricted T cell responses. Three patients maintained a partial clinical response or stable disease for more than 6 months. Median progression-free survival was 17.6 weeks, and median overall survival was 86.6 weeks from study entry with seven of nine patients surviving more than 12 months.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/imunologia , Glioma/terapia , Imunoterapia Ativa/métodos , Proteínas Inibidoras de Apoptose/imunologia , Peptídeos/imunologia , Linfócitos T/imunologia , Adulto , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Feminino , Glioma/imunologia , Glioma/mortalidade , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/metabolismo , Humanos , Imunidade Humoral , Proteínas Inibidoras de Apoptose/genética , Interferon gama/metabolismo , Masculino , Pessoa de Meia-Idade , Peptídeos/genética , Recidiva , Análise de Sobrevida , Survivina , Resultado do Tratamento , Vacinas de Subunidades
5.
Arch Immunol Ther Exp (Warsz) ; 62(5): 423-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25033772

RESUMO

The killer immunoglobulin-like receptor (KIR) genes KIR2DL4, KIR3DL2, and KIR3DP1 are present in virtually all humans. KIR2DL4 encodes a receptor present on uterine and decidual natural killer (NK) cells and some peripheral blood NK cells. Its only known ligand is the human leukocyte antigen-G molecule expressed on extravillous trophoblasts, and on tissues in some diseases. KIR3DL2 binds HLA-A*03 and HLA-A*11 as well as HLA-B*27 dimers, and microbial CpG DNA. KIR3DP1 is a pseudogene. During our immunogenetic studies we found two individuals, one from Lower Silesia district in Poland, and another from Western Ukraine, who were reproducibly negative for KIR2DL4 and KIR3DP1 genes, using three different PCR systems. Both individuals displayed very similar genotypes, possessing only KIR3DL3, KIR2DL3, KIR2DP1, KIR2DS1, and probably a rare variant of KIR2DL1. The Pole had also KIR3DL2, which the Ukrainian was apparently lacking. The Lower Silesia has been populated after the Second World War by a remarkable percentage with displaced people from Western Ukraine, which might contribute to genetic similarity of the two individuals described here.


Assuntos
Células Matadoras Naturais/imunologia , Receptores KIR2DL4/metabolismo , Receptores KIR3DL2/metabolismo , Idoso , Citotoxicidade Imunológica/genética , Genética Populacional , Genótipo , Antígenos HLA-A/metabolismo , Antígeno HLA-A3/metabolismo , Antígeno HLA-B27/metabolismo , Antígenos HLA-G/metabolismo , Humanos , Masculino , Polônia , Polimorfismo Genético , Ligação Proteica , Pseudogenes/genética , Receptores KIR/genética , Receptores KIR2DL4/genética , Receptores KIR3DL2/genética , Ucrânia , Adulto Jovem
6.
J Immunol ; 192(4): 1962-71, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24453253

RESUMO

Peptide splicing is a novel mechanism of production of peptides relying on the proteasome and involving the linkage of fragments originally distant in the parental protein. Peptides produced by splicing can be presented on class I molecules of the MHC and recognized by CTLs. In this study, we describe a new antigenic peptide, which is presented by HLA-A3 and comprises two noncontiguous fragments of the melanoma differentiation Ag gp100(PMEL17) spliced together in the reverse order to that in which they appear in the parental protein. Contrary to the previously described spliced peptides, which are produced by the association of fragments of 3-6 aa, the peptide described in this work results from the ultimate association of an 8-aa fragment with a single arginine residue. As described before, peptide splicing takes place in the proteasome by transpeptidation involving an acyl-enzyme intermediate linking one of the peptide fragment to a catalytic subunit of the proteasome. Interestingly, we observe that the peptide causing the nucleophilic attack on the acyl-enzyme intermediate must be at least 3 aa long to give rise to a spliced peptide. The spliced peptide produced from this reaction therefore bears an extended C terminus that needs to be further trimmed to produce the final antigenic peptide. We show that the proteasome is able to perform the final trimming step required to produce the antigenic peptide described in this work.


Assuntos
Melanoma/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína/fisiologia , Antígeno gp100 de Melanoma/genética , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Antígeno HLA-A3/genética , Antígeno HLA-A3/imunologia , Antígeno HLA-A3/metabolismo , Humanos , Melanoma/genética , Melanoma/imunologia , Fragmentos de Peptídeos/genética , Linfócitos T Citotóxicos/imunologia , Antígeno gp100 de Melanoma/imunologia , Antígeno gp100 de Melanoma/metabolismo
7.
J Immunol ; 190(7): 3216-24, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23440420

RESUMO

The human leukocyte Ag HLA-B27 (B27) is strongly associated with the spondyloarthritides. B27 can be expressed at the cell surface of APC as both classical ß2-microglobulin-associated B27 and B27 free H chain forms (FHC), including disulfide-bonded H chain homodimers (termed B27(2)). B27 FHC forms, but not classical B27, bind to KIR3DL2. HLA-A3, which is not associated with spondyloarthritis (SpA), is also a ligand for KIR3DL2. In this study, we show that B27(2) and B27 FHC bind more strongly to KIR3DL2 than other HLA-class I, including HLA-A3. B27(2) tetramers bound KIR3DL2-transfected cells more strongly than HLA-A3. KIR3DL2Fc bound to HLA-B27-transfected cells more strongly than to cells transfected with other HLA-class I. KIR3DL2Fc pulled down multimeric, dimeric, and monomeric FHC from HLA-B27-expressing cell lines. Binding to B27(2) and B27 FHC stimulated greater KIR3DL2 phosphorylation than HLA-A3. B27(2) and B27 FHC stimulated KIR3DL2CD3ε-transduced T cell IL-2 production to a greater extent than control HLA-class I. KIR3DL2 binding to B27 inhibited NK IFN-γ secretion and promoted greater survival of KIR3DL2(+) CD4 T and NK cells than binding to other HLA-class I. KIR3DL2(+) T cells from B27(+) SpA patients proliferated more in response to Ag presented by syngeneic APC than the same T cell subset from healthy and disease controls. Our results suggest that expansion of KIR3DL2-expressing leukocytes observed in B27(+) SpA may be explained by the stronger interaction of KIR3DL2 with B27 FHC.


Assuntos
Antígeno HLA-B27/metabolismo , Receptores KIR3DL2/metabolismo , Espondilite Anquilosante/imunologia , Espondilite Anquilosante/metabolismo , Antígenos/imunologia , Linhagem Celular , Sobrevivência Celular/imunologia , Células Cultivadas , Antígeno HLA-A3/imunologia , Antígeno HLA-A3/metabolismo , Antígeno HLA-B27/química , Antígeno HLA-B27/imunologia , Antígeno HLA-B35/imunologia , Antígeno HLA-B35/metabolismo , Antígeno HLA-B7/imunologia , Antígeno HLA-B7/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ligação Proteica , Multimerização Proteica , Receptores KIR3DL2/genética , Receptores KIR3DL2/imunologia , Espondilite Anquilosante/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
8.
Hum Vaccin Immunother ; 8(7): 987-1000, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22777092

RESUMO

Two major obstacles confronting HIV vaccine design have been the extensive viral diversity of HIV-1 globally and viral evolution driven by escape from CD8(+) cytotoxic T-cell lymphocyte (CTL)-mediated immune pressure. Regions of the viral genome that are not able to escape immune response and that are conserved in sequence and across time may represent the "Achilles' heel" of HIV and would be excellent candidates for vaccine development. In this study, T-cell epitopes were selected using immunoinformatics tools, combining HLA-A3 binding predictions with relative sequence conservation in the context of global HIV evolution. Twenty-seven HLA-A3 epitopes were chosen from an analysis performed in 2003 on 10,803 HIV-1 sequences, and additional sequences were selected in 2009 based on an expanded set of 43,822 sequences. These epitopes were tested in vitro for HLA binding and for immunogenicity with PBMCs of HIV-infected donors from Providence, Rhode Island. Validation of these HLA-A3 epitopes conserved across time, clades, and geography supports the hypothesis that epitopes such as these would be candidates for inclusion in our globally relevant GAIA HIV vaccine constructs.


Assuntos
Vacinas contra a AIDS/imunologia , Sequência Conservada , Epitopos de Linfócito T/imunologia , HIV-1/imunologia , Biologia Computacional/métodos , Epitopos de Linfócito T/metabolismo , Infecções por HIV/imunologia , Antígeno HLA-A3/imunologia , Antígeno HLA-A3/metabolismo , Humanos , Leucócitos Mononucleares/imunologia , Rhode Island
10.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 5): 447-54, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21543847

RESUMO

The structure of the human major histocompatability (MHC) class I molecule HLA-A*0301 (HLA-A3) in complex with a nonameric peptide (KLIETYFSK) has been determined by X-ray crystallography to 2.7 Šresolution. HLA-A3 is a predisposing allele for multiple sclerosis (MS), an autoimmune disease of the central nervous system. The KLIETYFSK peptide is a naturally processed epitope of proteolipid protein, a myelin protein and candidate target for immune-mediated myelin destruction in MS. Comparison of the structure of HLA-A3 with that of HLA-A2, an MHC class I molecule which is protective against MS, indicates that both MHC class I molecules present very similar faces for T-cell receptor recognition whilst differing in the specificity of their peptide-binding grooves. These characteristics may underlie the opposing (predisposing versus protective) associations that they exhibit both in humans and in mouse models of MS-like disease. Furthermore, subtle alterations within the peptide-binding groove of HLA-A3 and other A3-like MHC class I molecules, members of the so-called A3 superfamily, may be sufficient to alter their presentation of autoantigen peptides such as KLIETYFSK. This in turn may modulate their contribution to the associated risk of autoimmune disease.


Assuntos
Antígeno HLA-A3/química , Antígeno HLA-A3/metabolismo , Esclerose Múltipla/metabolismo , Proteolipídeos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Antígeno HLA-A2/química , Antígeno HLA-A2/metabolismo , Humanos , Modelos Moleculares , Peptídeos/química , Peptídeos/metabolismo , Proteolipídeos/química
11.
Nat Immunol ; 12(1): 45-53, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21151101

RESUMO

Cytotoxic T lymphocytes (CTLs) recognize peptides presented by HLA class I molecules on the cell surface. The C terminus of these CTL epitopes is considered to be produced by the proteasome. Here we demonstrate that the cytosolic endopeptidases nardilysin and thimet oligopeptidase (TOP) complemented proteasome activity. Nardilysin and TOP were required, either together or alone, for the generation of a tumor-specific CTL epitope from PRAME, an immunodominant CTL epitope from Epstein-Barr virus protein EBNA3C, and a clinically important epitope from the melanoma protein MART-1. TOP functioned as C-terminal trimming peptidase in antigen processing, and nardilysin contributed to both the C-terminal and N-terminal generation of CTL epitopes. By broadening the antigenic peptide repertoire, nardilysin and TOP strengthen the immune defense against intracellular pathogens and cancer.


Assuntos
Antígenos de Neoplasias/metabolismo , Epitopos de Linfócito T/metabolismo , Metaloendopeptidases/metabolismo , Linfócitos T Citotóxicos/metabolismo , Apresentação de Antígeno/genética , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Citotoxicidade Imunológica/genética , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Antígeno HLA-A3/metabolismo , Humanos , Células K562 , Metaloendopeptidases/genética , Metaloendopeptidases/imunologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Transgenes/genética
12.
Eur J Haematol ; 84(2): 145-53, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19912313

RESUMO

BACKGROUND: The haemochromatosis mutation C282Y occurred once in a person who lived in Ireland or Scandinavia and carried either human leucocyte antigen (HLA)-A3-B7 or A3-B14. With time, recombinations are believed to have taken place introducing new HLA haplotypes. This evolution is mainly unknown. In this study, we tried to find a founder, possible recombination events and effect on the phenotype in descendants. SETTING: A Swedish mountain population close to Norway, n = 3529, population density <1/km(2). METHODS: Retrospective genealogy study of HLA haplotypes followed by extended haplotype studies. RESULTS: There were 34 probands (22 men, 12 women) where 31 (91%) shared a common founder origin 12 generations ago. The A3-B14 haplotype was the most common, 39%, in strong linkage disequilibrium (P < 0.0005) with controls, followed by A3-B7, 20% (P < 0.005), probably resulting from a centromeric recombination replacing the B14 allele with the common B7. Possible telomeric recombinations took place close to HLA-A and introduced the haplotypes AW19-B7 (n = 4), AW19-B27 (2), A1-B17 (5) and A2-B12 (4) supported by pedigree studies. Male homozygotes with two copies of HLA-A3 had significantly (P 0.001) higher mean serum ferritin values than those with one, and liver damage (fibrosis and cirrhosis) was also more common (P < 0.001) than in a population with a recombinant (A1-B8) haplotype. CONCLUSIONS: A3-B14 may well be the ancestral haplotype with A3B7, the result of centromeric recombinations introducing the common B7 allele. Telomeric recombinations were more common than expected. The ancestral HLA-A3 haplotype may be associated with a more severe phenotypic expression.


Assuntos
Efeito Fundador , Antígeno HLA-A3/genética , Hemocromatose/genética , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Recombinação Genética , Alelos , Substituição de Aminoácidos , Família , Feminino , Seguimentos , Antígeno HLA-A3/metabolismo , Haplótipos , Hemocromatose/metabolismo , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/metabolismo , Homozigoto , Humanos , Desequilíbrio de Ligação , Masculino , Proteínas de Membrana/metabolismo , Linhagem , Suécia
13.
J Immunol ; 181(9): 6010-9, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18941190

RESUMO

Stem cell transplantation across HLA barriers may trigger NK cell-mediated graft-vs-leukemia effects leading to improved survival for patients with hematological malignancies. However, the genetic algorithm based on killer cell Ig-like receptor (KIR) and HLA genes used to predict NK cell alloreactivity have yielded discrepant results. Accordingly, it has been difficult to define transplantation settings that favor NK cell alloreactivity. In this study, we have used multiparameter flow cytometry to simultaneously analyze the cell surface expression of all four major inhibitory KIR and CD94/NKG2A to determine the size of the alloreactive NK cell repertoires in 31 individuals homozygous for the group A KIR haplotype. We observed a vast variability in the frequencies of cells with an alloreactive potential, ranging from 0 to 62% of the total NK cell population depending on which, and how many, KIR ligands were missing in theoretical recipients. This analysis required a functional examination of KIR3DL2-single positive NK cells, showing that this subset was hyporesponsive in individuals harboring the cognate ligands HLA-A3/A11. The results provide new insights into the variability of the functional alloreactive NK cell repertoire and have implications for donor selection in hematopoietic stem cell transplantation and adoptive NK cell-based immunotherapy.


Assuntos
Haplótipos , Teste de Histocompatibilidade , Homozigoto , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Receptores KIR3DL2/genética , Anticorpos Monoclonais/metabolismo , Testes Imunológicos de Citotoxicidade , Genótipo , Efeito Enxerto vs Leucemia/genética , Efeito Enxerto vs Leucemia/imunologia , Antígenos HLA-A/metabolismo , Antígeno HLA-A11 , Antígeno HLA-A3/metabolismo , Teste de Histocompatibilidade/métodos , Humanos , Células K562 , Células Matadoras Naturais/metabolismo , Ligantes , Subfamília C de Receptores Semelhantes a Lectina de Células NK/biossíntese , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores KIR3DL2/biossíntese , Receptores KIR3DL2/imunologia , Receptores KIR3DL2/metabolismo , Transplante de Células-Tronco
14.
Scand J Immunol ; 64(4): 444-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16970688

RESUMO

Target cell resistance against natural killer (NK) cell-mediated cytotoxicity obstructs NK cell-based immunotherapy of leukaemia. Several mechanisms of resistance have been described. Because of lack of simple assays for analysing these mechanisms, their relative impact on a given effector-target pair is mostly unknown. We here analysed the combination of the Granzyme B (GrB) enzyme-linked immunospot assay (ELISPOT) for the assessment of NK cell reactivity and cytotoxicity assays to estimate target cell escape mechanisms. Target cell recognition failure leads to negative GrB ELISPOT results, whereas target cell resistance shows positive GrB ELISPOT results in the absence of cytotoxicity. We confronted NK cells with the sensitive target cell line K562, and with the resistant cell lines ML2, SupB15 and Raji. ML2 cells sufficiently activated GrB-release whilst being resistant against cytotoxic granules of NK cells. Partial resistance of Raji results from the interaction of HLA class I with inhibitory killer immunglobulin-like receptors (KIR) on the NK cells. Failure of target recognition by HLA class I-KIR interaction, lacking ligands to stimulatory NK cell receptors and partial resistance to cytotoxic granules all contributed to resistance of SupB15. In conclusion, revealing the mechanisms of resistance against NK cell-mediated cytotoxicity may allow improving the results of NK-based immunotherapy.


Assuntos
Testes Imunológicos de Citotoxicidade/métodos , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Linhagem Celular Tumoral , Células Clonais , Granzimas , Antígeno HLA-A3/biossíntese , Antígeno HLA-A3/genética , Antígeno HLA-A3/metabolismo , Humanos , Imunidade Inata/genética , Células K562 , Células Matadoras Naturais/enzimologia , Células Matadoras Naturais/metabolismo , Leucemia/enzimologia , Leucemia/imunologia , Leucemia/patologia , Receptores Imunológicos/biossíntese , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores KIR , Serina Endopeptidases/análise
15.
Int J Cancer ; 119(10): 2476-80, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16858680

RESUMO

The clonotypic T-cell receptor (TCR) is a potential target antigen for specific immunotherapy of cutaneous T-cell lymphoma (CTCL). We identified T-cell epitopes from the rearranged TCR beta chain of the malignant T-cell population by the "reverse immunology" approach. Peptide-specific T-cell lines were generated against predicted epitopes and tested for the recognition of tumor cells and cells transfected with the full-length DNA coding for TCRV beta chain. Two peptides derived from the clonotypic TCRVbeta of a HLA-A2 positive patient could induce peptide-specific T cells from peripheral blood mononuclear cells of healthy donors and the patient as assessed by IFN-gamma ELISpot assay. Furthermore, the reactive CTLs efficiently recognized autologous Sézary tumor cells, as well as HLA-A2 positive 293 cells transfected with recombinant plasmid expressing the corresponding TCRVbeta29 protein. Similar results were obtained in a HLA-A3+ patient for TCRVbeta7-Jbeta2.7. In conclusion, our experiments show that the TCR beta chain harbors epitopes suitable as targets for specific vaccination which might be a promising approach for the specific immunotherapy of cutaneous T-cell lymphoma patients.


Assuntos
Epitopos/imunologia , Linfoma Cutâneo de Células T/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Neoplasias Cutâneas/imunologia , Regulação Neoplásica da Expressão Gênica , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/metabolismo , Humanos , Imunoterapia/métodos , Leucócitos Mononucleares/imunologia , Linfoma Cutâneo de Células T/terapia , Peptídeos/imunologia , Síndrome de Sézary/imunologia , Neoplasias Cutâneas/terapia
16.
Cancer Immunol Immunother ; 54(4): 359-71, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15378283

RESUMO

The development of peptide-based vaccines that are useful in the therapeutic treatment of melanoma and other cancers ultimately requires the identification of a sufficient number of antigenic peptides so that most individuals, regardless of their major histocompatibility complex (MHC)-encoded class I molecule phenotype, can develop a cytotoxic T lymphocyte (CTL) response against one or more peptide components of the vaccine. While it is relatively easy to identify antigenic peptides that are presented by the most prevalent MHC class I molecules in the population, it is problematic to identify antigenic peptides that are presented by MHC class I molecules that have less frequent expression in the population. One manner in which this problem can be overcome is by taking advantage of known MHC class I supertypes, which are groupings of MHC class I molecules that bind peptides sharing a common motif. We have developed a mass spectrometric approach which can be used to determine if an antigenic peptide is naturally processed and presented by any given MHC class I molecule. This approach has been applied to the A3 supertype, and the results demonstrate that some, but not all, A3 supertype family-associated peptides can associate with all A3 supertype family members. The approach also demonstrates the shared nature of several newly identified peptide antigens. The use of this technology negates the need to test peptides for their ability to stimulate CTL responses in those cases where the peptide is not naturally processed and bound to the target MHC class I molecule of interest, thus allowing resources to be focused on the most promising vaccine candidates.


Assuntos
Epitopos/imunologia , Antígeno HLA-A3/imunologia , Melanoma/imunologia , Fragmentos de Peptídeos/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T Citotóxicos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Cromatografia Líquida de Alta Pressão , Epitopos/isolamento & purificação , Epitopos/metabolismo , Análise de Fourier , Antígeno HLA-A3/metabolismo , Humanos , Espectrometria de Massas , Melanoma/metabolismo , Melanoma/patologia , Fragmentos de Peptídeos/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
17.
J Reprod Immunol ; 62(1-2): 3-15, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15288176

RESUMO

During pregnancy, the non-classical major histocompatibility complex (MHC) class I HLA-G molecule is specifically expressed in trophoblast cells at the materno-fetal interface and may exert a local control of the immune response against viral infections. Human cytomegalovirus (HCMV) infection, which is the major cause of congenital defects, encodes multiple glycoproteins (US2, US3, US6, US10 and US11) that interrupt the MHC class I pathway of antigen presentation. The effect of some of these unique short (US) proteins on HLA-G expression has been previously studied, but little is known about the modulation of HLA-G cell surface expression during the course of HCMV infection which ensures expression of all of these US proteins. Using flow cytometry analysis, HLA-G cell surface expression was evaluated in HCMV-infected U373-HLA-G transfectant cells and compared with the modulation of the endogenous classical HLA-A2 molecules. The results indicated that HCMV infection down-modulated HLA-G cell surface expression, but later after infection and to a lesser extent than HLA-A2. Using various HLA-G/HLA-A2 chimeras, we showed that the unique structure of HLA-G cytoplasmic tail was partly involved in the resistance of HLA-G to viral down-modulation. Such limited down-modulation of HLA-G may have functional consequences in term of innate immunity against congenital HCMV infection.


Assuntos
Infecções por Citomegalovirus/metabolismo , Regulação para Baixo , Antígenos HLA/metabolismo , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citoplasma/imunologia , Citoplasma/metabolismo , Citoplasma/virologia , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Feminino , Antígenos HLA/análise , Antígenos HLA/genética , Antígeno HLA-A2/análise , Antígeno HLA-A2/genética , Antígeno HLA-A3/análise , Antígeno HLA-A3/genética , Antígenos HLA-G , Antígenos de Histocompatibilidade Classe I/análise , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Dados de Sequência Molecular , Gravidez , Deleção de Sequência , Proteínas Virais/análise , Proteínas Virais/genética , Proteínas Virais/metabolismo
18.
J Virol ; 78(13): 7069-78, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15194783

RESUMO

Numerous studies now support that human immunodeficiency virus type 1 (HIV-1) evolution is influenced by immune selection pressure, with population studies showing an association between specific HLA alleles and mutations within defined cytotoxic T-lymphocyte epitopes. Here we combine sequence data and functional studies of CD8 T-cell responses to demonstrate that allele-specific immune pressures also select for mutations flanking CD8 epitopes that impair antigen processing. In persons expressing HLA-A3, we demonstrate consistent selection for a mutation in a C-terminal flanking residue of the normally immunodominant Gag KK9 epitope that prevents its processing and presentation, resulting in a rapid decline in the CD8 T-cell response. This single amino acid substitution also lies within a second HLA-A3-restricted epitope, with the mutation directly impairing recognition by CD8 T cells. Transmission of the mutation to subjects expressing HLA-A3 was shown to prevent the induction of normally immunodominant acute-phase responses to both epitopes. However, subsequent in vivo reversion of the mutation was coincident with delayed induction of new CD8 T-cell responses to both epitopes. These data demonstrate that mutations within the flanking region of an HIV-1 epitope can impair recognition by an established CD8 T-cell response and that transmission of these mutations alters the acute-phase CD8(+) T-cell response. Moreover, reversion of these mutations in the absence of the original immune pressure reveals the potential plasticity of immunologically selected evolutionary changes.


Assuntos
Substituição de Aminoácidos , Apresentação de Antígeno , Evolução Molecular , Produtos do Gene gag/genética , Antígenos HIV/genética , HIV-1/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/genética , Sequência de Aminoácidos , Epitopos de Linfócito T/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , Antígeno HLA-A3/metabolismo , Humanos , Epitopos Imunodominantes/imunologia , Dados de Sequência Molecular , Seleção Genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...