Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 338
Filter
1.
J Reprod Immunol ; 156: 103817, 2023 03.
Article in English | MEDLINE | ID: mdl-36753935

ABSTRACT

Immune system aberrations are suggested to be an important factor in the pathophysiology of unexplained secondary recurrent pregnancy loss (sRPL). The objective was to investigate if the sex ratio of the firstborn child in sRPL patients differs from the background population and whether the sex of the firstborn child has a negative impact on the pregnancy prognosis alone and/or in combination with carriage of male-specific minor histocompatibility (H-Y) restricting HLA class II alleles. From January 2016 to October 2022, 582 patients with unexplained RPL were admitted to the RPL Center of Western Denmark and continuously followed-up. HLA-DRB1 and -DQB1 typing was performed as part of the routine diagnostic work-up. In sRPL patients, a history of a firstborn boy was significantly more frequent than in the Danish background population and was associated with significantly lower odds of a successful reproductive outcome in the first pregnancy after admission compared to a firstborn girl (OR=0.41, 95% CI: 0.20-0.83, p = 0.014). The odds of a successful reproductive outcome were enhanced in patients carrying ≥ 1 H-Y-restricting HLA class II alleles with a first-born girl compared to a firstborn boy (OR=3.33, 95% CI: 1.40-7.88, p = 0.005), while no difference in successful reproductive outcome was seen in sRPL patients not carrying these alleles (OR=1.20, 95% CI: 0.33-4.43, p = 0.781). The sex ratio of children born after RPL was similar to the Danish background population. These findings confirm previous findings and suggests that a harmful immune response triggered by H-Y-antigen exposure during a previous pregnancy in preconditioned women may cause sRPL.


Subject(s)
Abortion, Habitual , H-Y Antigen , Female , Humans , Male , Pregnancy , Alleles , H-Y Antigen/genetics , Histocompatibility , HLA-DRB1 Chains/genetics , Prognosis , Histocompatibility Antigens Class II/immunology
2.
Anim Biotechnol ; 31(6): 561-565, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31198087

ABSTRACT

Polled intersex syndrome (PIS), a physiological defect associates polledness and intersexuality, will directly affect the reproduction and other phenotypic traits of goats, thus, it is a hazard to the development of the goat industry. Recent studies have revealed that goat PIS was caused by an 11.7-kb deletion located in chromosome 1q43, but its DNA detection method was little reported. Herein, a total of 503 goats from three goat breeds were used to successfully establish a simple, rapid, accurate and effective method for the detection of goat PIS mutation. Furthermore, based on this assay, the associations between the PIS mutation and growth traits were analyzed in Guanzhong dairy goats. These findings would provide the potential practical application for molecular breeding to accelerate the development of the goat industry.


Subject(s)
Disorders of Sex Development , Goats , Animals , China , Dairying , Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Disorders of Sex Development/veterinary , Female , Goats/genetics , Goats/growth & development , H-Y Antigen/genetics , Mutation/genetics , Polymerase Chain Reaction
3.
J Immunol ; 195(2): 717-25, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26048147

ABSTRACT

Naturally derived regulatory T cells (Tregs) may prevent graft-versus-host disease (GVHD) while preserving graft-versus-leukemia (GVL) activity. However, clinical application of naturally derived regulatory T cells has been severely hampered by their scarce availability and nonselectivity. To overcome these limitations, we took alternative approaches to generate Ag-specific induced Tregs (iTregs) and tested their efficacy and selectivity in the prevention of GVHD in preclinical models of bone marrow transplantation. We selected HY as a target Ag because it is a naturally processed, ubiquitously expressed minor histocompatibility Ag (miHAg) with a proven role in GVHD and GVL effect. We generated HY-specific iTregs (HY-iTregs) from resting CD4 T cells derived from TCR transgenic mice, in which CD4 cells specifically recognize HY peptide. We found that HY-iTregs were highly effective in preventing GVHD in male (HY(+)) but not female (HY(-)) recipients using MHC II-mismatched, parent→F1, and miHAg-mismatched murine bone marrow transplantation models. Interestingly, the expression of target Ag (HY) on the hematopoietic or nonhematopoietic compartment alone was sufficient for iTregs to prevent GVHD. Furthermore, treatment with HY-iTregs still preserved the GVL effect even against pre-established leukemia. We found that HY-iTregs were more stable in male than in female recipients. Furthermore, HY-iTregs expanded extensively in male but not female recipients, which in turn significantly reduced donor effector T cell expansion, activation, and migration into GVHD target organs, resulting in effective prevention of GVHD. This study demonstrates that iTregs specific for HY miHAgs are highly effective in controlling GVHD in an Ag-dependent manner while sparing the GVL effect.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease/prevention & control , H-Y Antigen/immunology , Leukemia/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Female , Gene Expression , Graft vs Leukemia Effect , H-Y Antigen/genetics , Histocompatibility Testing , Leukemia/genetics , Leukemia/immunology , Leukemia/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Sex Factors , Survival Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation , Transplantation, Homologous , Whole-Body Irradiation
4.
Immunology ; 144(1): 149-57, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25039245

ABSTRACT

A role for complement, particularly the classical pathway, in the regulation of immune responses is well documented. Deficiencies in C1q or C4 predispose to autoimmunity, while deficiency in C3 affects the suppression of contact sensitization and generation of oral tolerance. Complement components including C3 have been shown to be required for both B-cell and T-cell priming. The mechanisms whereby complement can mediate these diverse regulatory effects are poorly understood. Our previous work, using the mouse minor histocompatibility (HY) model of skin graft rejection, showed that both C1q and C3 were required for the induction of tolerance following intranasal peptide administration. By comparing tolerance induction in wild-type C57BL/6 and C1q-, C3-, C4- and C5-deficient C57BL/6 female mice, we show here that the classical pathway components including C3 are required for tolerance induction, whereas C5 plays no role. C3-deficient mice failed to generate a functional regulatory T (Treg) -dendritic cell (DC) tolerogenic loop required for tolerance induction. This was related to the inability of C3-deficient DC to up-regulate the arginine-consuming enzyme, inducible nitric oxide synthase (Nos-2), in the presence of antigen-specific Treg cells and peptide, leading to reduced Treg cell generation. Our findings demonstrate that the classical pathway and C3 play a critical role in the peptide-mediated induction of tolerance to HY by modulating DC function.


Subject(s)
Complement System Proteins/genetics , Dendritic Cells/immunology , H-Y Antigen/immunology , Immune Tolerance/drug effects , Peptides/pharmacology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Animals , Complement System Proteins/immunology , Dendritic Cells/cytology , Female , H-Y Antigen/genetics , Immune Tolerance/genetics , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Peptides/immunology , T-Lymphocytes, Regulatory/cytology
5.
PLoS One ; 9(3): e91274, 2014.
Article in English | MEDLINE | ID: mdl-24646895

ABSTRACT

BACKGROUND: Sex difference is an established risk factor for hematopoietic stem cell transplantation (HSCT)-related complications like graft versus host disease (GVHD). CD8pos cytotoxic T cells specific for Y chromosome-encoded minor Histocompatibility antigens (HY) play an important role therein. Prior to HSC donation, female donors may encounter HY antigens through fetomaternal or transmaternal cell flow, potentially leading to the induction of HY-specific cytotoxic or regulatory immune responses. Whether HY priming occurs independent of parity, and whether HY priming is dependent on the presence of male microchimerism, is as yet unknown. METHODS: We investigated the presence of HY-specific regulatory T cells (Treg) and male microchimerism in 45 healthy women with a fully documented pregnancy and family history. HY peptide-induced linked suppression, a commonly reported functional feature of CD4pos and CD8pos Treg, was measured by trans vivo Delayed Type Hypersensitivity testing. As source of HY antigens, male microchimerism was analyzed by real-time PCR and defined by the presence of male DNA in at least one purified leukocyte cell type. RESULTS: HLA class I or class II restricted HY-specific Treg were detected in 26/42 (62%) women eligible for analysis. The prevalence of HY-specific Treg was significantly higher in women who had never given birth to sons than in women with male offspring (p = 0.004). Male microchimerism could be detected in 24 out of 45 (53%) women but did not correlate with the presence of HY specific Treg. CONCLUSIONS: HY-specific Treg in women with male offspring have been described previously. Here we show for the first time that, in fact, HY specific Treg are more common in nulliparous women and in parous women with female offspring. Their presence is independent of the presence of male microchimerism. Whether HY-specific Treg presence in female stem cell grafts might decrease the GVHD incidence in male HSCT recipients needs to be investigated.


Subject(s)
H-Y Antigen/immunology , Immune Tolerance , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Amino Acid Sequence , Animals , Chimerism , Female , Gene Expression , H-Y Antigen/genetics , Humans , Male , Mice , Molecular Sequence Data , Pregnancy , Sex Factors , T-Lymphocytes, Cytotoxic/chemistry , T-Lymphocytes, Regulatory/chemistry
6.
Biol Blood Marrow Transplant ; 20(1): 26-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141010

ABSTRACT

The clinical success of allogeneic T cell therapy for cancer relies on the selection of antigens that can effectively elicit antitumor responses with minimal toxicity toward nonmalignant tissues. Although minor histocompatibility antigens (MiHA) represent promising targets, broad expression of these antigens has been associated with poor responses and T cell dysfunction that may not be prevented by targeting MiHA with limited expression. In this study, we hypothesized that antitumor activity of MiHA-specific CD8 T cells after allogeneic bone marrow transplantation (BMT) is determined by the distribution of antigen relative to the site of tumor growth. To test this hypothesis, we utilized the clinically relevant male-specific antigen HY and studied the fate of adoptively transferred, HY-CD8(+) T cells (HY-CD8) against a HY-expressing epithelial tumor (MB49) and pre-B cell leukemia (HY-E2APBX ALL) in BMT recipients. Transplants were designed to produce broad HY expression in nonhematopoietic tissues (female → male BMT, [F → M]), restricted HY expression in hematopoietic tissues (male → female BMT, [M → F]) tissues, and no HY tissue expression (female → female BMT, [F → F]). Broad HY expression induced poor responses to MB49 despite sublethal graft-versus-host disease and accumulation of HY-CD8 in secondary lymphoid tissues. Antileukemia responses, however, were preserved. In contrast, restriction of HY expression to hematopoietic tissues restored MB49 responses but resulted in a loss of antileukemia responses. We concluded that target alloantigen expression in the same compartment of tumor growth impairs CD8 responses to both solid and hematologic tumors.


Subject(s)
Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect , H-Y Antigen/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Adoptive Transfer , Alleles , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression/immunology , Graft vs Host Disease/genetics , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , H-Y Antigen/genetics , Humans , Immunophenotyping , Lymphocyte Depletion , Male , Mice , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/pathology , Survival Analysis
7.
Transpl Immunol ; 29(1-4): 138-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24161680

ABSTRACT

Alloreactive T-cell responses directed against minor histocompatibility (H) antigens, which arise from diverse genetic disparities between donor and recipient outside the MHC, are an important cause of rejection of MHC-matched grafts. Because clinically significant responses appear to be directed at only a few antigens, the selective deletion of naïve T cells recognizing donor-specific, immunodominant minor H antigens in recipients before transplantation may be a useful tolerogenic strategy. We have previously demonstrated that peptide-MHC class I tetramers coupled to a toxin can efficiently eliminate specific TCR-transgenic T cells in vivo. Here, using the minor histocompatibility antigen HY as a model, we investigated whether toxic tetramers could inhibit the subsequent priming of the two H2-D(b)-restricted, immunodominant T-cell responses by deleting precursor CTL. Immunization of female mice with male bone marrow elicited robust CTL activity against the Uty and Smcy epitopes, with Uty constituting the major response. As hypothesized, toxic tetramer administration prior to immunization increased survival of cognate peptide-pulsed cells in an in vivo CTL assay, and reduced the frequency of corresponding T cells. However, tetramer-mediated decreases in either T-cell population magnified CTL responses against the non-targeted epitope, suggesting that D(b)-Uty(+) and D(b)-Smcy(+) T cells compete for a limited common resource during priming. Toxic tetramers conceivably could be used in combination to dissect manipulate CD8(+) T-cell immunodominance hierarchies, and to prevent the induction of donor-specific, minor H antigen CTL responses in allotransplantation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , H-Y Antigen/immunology , Histocompatibility Antigens Class I/immunology , Immunotoxins/immunology , Lymphocyte Depletion/methods , Peptides/immunology , Allografts , Animals , Bone Marrow Transplantation , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , H-Y Antigen/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/pharmacology , Immunotoxins/genetics , Immunotoxins/pharmacology , Male , Mice , Mice, Transgenic , Peptides/genetics , Peptides/pharmacology
8.
Chimerism ; 4(1): 18-9, 2013.
Article in English | MEDLINE | ID: mdl-23262442

ABSTRACT

It is well accepted that pregnancy establishes microchimerism. Lately, transmaternal passage of cells from elder siblings has been suggested as possible source of non-fetal microchimerism in nulliparous women. Recently, we identified both non-maternal microchimerism and minor H antigen specific cellular immune responses against these microchimeric cells in umbilical cord blood. The identification of the latter microchimeric cells from birth onwards explains the high incidence of microchimerism in healthy people and in people with different diseases. The circulating microchimeric cells of different origin can induce a wealth of antigen specific responses. Thus, nobody is born naïve: we are all microchimera's.


Subject(s)
Autoimmune Diseases/genetics , Chimerism , Fetal Blood/immunology , Maternal-Fetal Exchange , Autoimmune Diseases/etiology , Female , Fetus , H-Y Antigen/genetics , H-Y Antigen/immunology , Humans , Male , Maternal-Fetal Exchange/genetics , Maternal-Fetal Exchange/immunology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Pregnancy
9.
Methods Mol Biol ; 723: 81-104, 2011.
Article in English | MEDLINE | ID: mdl-21370061

ABSTRACT

Recombinant antigen arrays represent a new frontier in parallel analysis of multiple immune response profiles requiring only minute blood samples. In this article, we review the benefits and pitfalls of recombinant antigen microarrays developed for multiplexed antibody quantification. In particular, we describe the development of antigen arrays presenting a set of Y chromosome-encoded antigens, called H-Y antigens. These H-Y antigens are immunologically recognized as minor histocompatibility antigens (mHA) following allogeneic blood and organ transplantation. Clinically relevant B-cell responses against H-Y antigens have been demonstrated in male patients receiving female hematopoietic stem cell grafts and are associated with chronic graft versus host development. This chapter discusses our recombinant antigen microarray methods to measure these clinically relevant allo-antibodies.


Subject(s)
H-Y Antigen/immunology , Isoantibodies/blood , Protein Array Analysis/methods , Recombinant Proteins/immunology , Serologic Tests/methods , Bacteria/cytology , Bacteria/genetics , Cell Culture Techniques , Epitopes/immunology , Female , H-Y Antigen/genetics , H-Y Antigen/isolation & purification , Humans , Isoantibodies/immunology , Male , Printing , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
10.
Hum Gene Ther ; 22(1): 44-54, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20649484

ABSTRACT

Graft-versus-host disease is still a major complication in stem-cell transplantation. In HLA-identical stem-cell transplantation, mismatches in minor histocompatibility antigens contribute to the development of graft-versus-host disease. As treatment of graft-versus-host disease with immunosuppressive drugs potentially also reduces the graft-versus-leukemia responses, antigen-specific tolerance induction may be used to reduce graft-versus-host disease while leaving the graft-versus-leukemia responses intact. In mouse models, induction of antigen-specific tolerance using IgG-antigen-transduced B cells can prevent and ameliorate autoimmunity. In this study, the principle of the in vivo animal model has been applied to a human in vitro model wherein induction of tolerance against the HLA class II-restricted male-specific minor histocompatibility antigen RPS4Y1 has been analyzed. T cells precultured in the presence of B cells expressing the IgG-coupled HY antigen showed reduced responsiveness selectively against the minor HY antigen RPS4Y1 in different in vitro protocols. These first observations serve as a basis for further studying and understanding the tolerizing potential of IgG constructs with human T cells.


Subject(s)
Genetic Therapy , H-Y Antigen/therapeutic use , Immunoglobulin G/therapeutic use , Stem Cell Transplantation , T-Lymphocytes/immunology , Animals , B-Lymphocytes/immunology , Cell Line , Female , Graft vs Host Disease/immunology , H-Y Antigen/genetics , H-Y Antigen/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immune Tolerance/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Male , Mice , Minor Histocompatibility Antigens/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/metabolism
11.
J Immunol ; 185(3): 1419-28, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20592282

ABSTRACT

Nonvirus-specific bystander CD8 T cells bathe in an inflammatory environment during viral infections. To determine whether bystander CD8 T cells are affected by these environments, we examined P14, HY, and OT-I TCR transgenic CD8 T cells sensitized in vivo by IFN-alphabeta-inducing viral infections or by polyinosinic:polycytidylic acid. These sensitized cells rapidly exerted effector functions, such as IFN-gamma production and degranulation, on contact with their high-affinity cognate Ag. Sensitization required self-MHC I and indirect effects of IFN-alphabeta, which together upregulated the T-box transcription factor Eomesodermin, potentially enabling the T cells to rapidly transcribe CTL effector genes and behave like memory cells rather than naive T cells. IL-12, IL-15, IL-18, and IFN-gamma were not individually required for sensitization to produce IFN-gamma, but IL-15 was required for upregulation of granzyme B. These experiments indicate that naive CD8 T cells receive signals from self-MHC and IFN-alphabeta and that, by this process, CD8 T cell responses to viral infection can undergo distinct differentiation pathways, depending on the timing of Ag encounter during the virus-induced IFN response.


Subject(s)
Autoantigens/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Histocompatibility Antigens Class I/physiology , Interferon-alpha/physiology , Interferon-beta/physiology , Signal Transduction/immunology , Animals , Arenaviridae Infections/immunology , Bystander Effect/immunology , CD8-Positive T-Lymphocytes/virology , Female , H-Y Antigen/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pichinde virus/immunology , Resting Phase, Cell Cycle/immunology , Up-Regulation/immunology
12.
Hum Reprod ; 25(6): 1543-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20395221

ABSTRACT

BACKGROUND: The chance of a live birth after a diagnosis of secondary recurrent miscarriage (SRM) is reduced in patients who, prior to the miscarriages, gave birth to a boy and carry HLA class II alleles that efficiently present male-specific (H-Y) antigens to the immune system. Information about obstetric complications in births prior and subsequent to the SRM diagnosis is limited. The relations between maternal carriage of H-Y-restricting HLA, fetal sex, obstetric complications and prognosis are unknown. METHODS: Women with unexplained SRM referred to the Danish Recurrent Miscarriage Clinic between 1986 and 2006 (n = 358) were included; 213 gave birth after the diagnosis. Controls, retrieved from the Danish National Birth Registry, were all women with singleton birth of parity 0, 1982-2005 (n = 608,068) and parity 1, 1986-2008 (n = 510,264). Cross-linkage to the National Discharge Registry identified birth complication diagnoses related to the relevant births among patients and controls. RESULTS: The sex ratio was 1.49 in births prior to SRM and 0.76 in birth after SRM (P < 0.0001). For SRM patients with only late miscarriages (>10 weeks gestation), the corresponding sex ratios were 2.31 and 0.21. Compared with the control groups, obstetric complications were more frequent both before (39% versus 24% P

Subject(s)
Abortion, Habitual/immunology , H-Y Antigen/immunology , HLA Antigens/immunology , Histocompatibility, Maternal-Fetal/immunology , Pregnancy Complications/immunology , Abortion, Habitual/genetics , Alleles , Denmark , Female , H-Y Antigen/genetics , HLA Antigens/genetics , Histocompatibility, Maternal-Fetal/genetics , Humans , Infant, Newborn , Live Birth , Male , Pregnancy , Pregnancy Complications/genetics , Registries , Risk Factors , Statistics, Nonparametric
13.
Blood ; 115(23): 4923-33, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20203263

ABSTRACT

T-cell immunotherapy that targets minor histocompatibility (H) antigens presented selectively by recipient hematopoietic cells, including leukemia, could prevent and treat leukemic relapse after hematopoietic cell transplantation without causing graft-versus-host disease. To provide immunotherapy that can be applied to a majority of transplantation recipients, it is necessary to identify leukemia-associated minor H antigens that result from gene polymorphisms that are balanced in the population and presented by common human leukocyte antigen alleles. Current approaches for deriving minor H antigen-specific T cells, which provide essential reagents for the molecular identification and characterization of the polymorphic genes that encode the antigens, rely on in vivo priming and are often unsuccessful. We show that minor H antigen-specific cytotoxic T lymphocyte precursors are found predominantly in the naive CD8(+) T-cell subset and provide an efficient strategy for in vitro priming of native T cells to generate T cells to a broad diversity of minor H antigens presented with common human leukocyte antigen alleles. We used this approach to derive a panel of stable cytotoxic T lymphocyte clones for discovery of genes that encode minor H antigens and identify a novel antigen expressed on acute myeloid leukemia stem cells and minimally in graft-versus-host disease target tissues.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , H-Y Antigen/genetics , H-Y Antigen/immunology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Neoplastic Stem Cells/immunology , Cell Line , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid, Acute/therapy , Male , Transplantation, Homologous
14.
Transfusion ; 50(6): 1295-302, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20088839

ABSTRACT

BACKGROUND: The Dombrock (Do) blood group system consists of five distinct antigens: Do(a), Do(b), Gy(a), Hy, and Jo(a). Our finding of a patient whose plasma contained a Do-related alloantibody suggested the presence of a sixth antigen. STUDY DESIGN AND METHODS: Standard hemagglutination, flow cytometry, and polymerase chain reaction (PCR)-based methods were used throughout. Protein homology modeling was used to map the amino acid change on the protein structure. RESULTS: The patient's red blood cells (RBCs) typed as Do(a-b-), Hy+(w), Jo(a+(w)), and Gy(a+(w)). The patient's plasma agglutinated RBCs with common Dombrock phenotypes. Reactivity with Hy- and Jo(a-) RBC samples was weak, and Gy(a-) RBC samples were nonreactive. DNA analysis showed the patient to be DO*793A (DO*A/DO*A), DO*323G, and DO*350C, which predicts the Do(a+b-), Hy+, and Jo(a+) phenotype, and revealed a homozygous single-nucleotide change of 547T>G in Exon 2 that is predicted to change tyrosine at Amino Acid Position 183 to aspartic acid. This missense substitution introduced a BtgZI restriction enzyme site. The sequence data were confirmed with a PCR-restriction fragment length polymorphism assay and revealed that the patient's parents and children were heterozygous DO*547T/G. Homology modeling predicted that the 183Tyr substitution by Asp altered the Cys182 environment and influenced the formation and/or stability of the Cys182-Cys231 disulfide bond. CONCLUSION: The patient's DO genes have a single-nucleotide change, which leads to the absence of the high-prevalence antigen DOYA. The absence of this antigen is associated with 183Asp and silencing of Do(a) and weakening of Gy(a), Hy, and Jo(a) antigens.


Subject(s)
ADP Ribose Transferases/biosynthesis , Blood Group Antigens/biosynthesis , Erythrocytes/metabolism , Gene Expression Regulation , H-Y Antigen/biosynthesis , Membrane Proteins/biosynthesis , Mutation, Missense , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Adult , Amino Acid Substitution , Blood Group Antigens/genetics , Disulfides/immunology , Disulfides/metabolism , Female , H-Y Antigen/genetics , H-Y Antigen/immunology , Homozygote , Humans , Isoantibodies/chemistry , Isoantibodies/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Polymorphism, Restriction Fragment Length/genetics , Polymorphism, Restriction Fragment Length/immunology
15.
Hum Mol Genet ; 18(9): 1684-91, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19223392

ABSTRACT

Healthy females, pregnant with a boy, generate immune responses against male-specific minor histocompatibility (HY) antigens. The clinical importance of these responses is evident in stem cell transplantation. Birth of a boy prior to a series of miscarriages reduces the chance of a subsequent live birth. This study explores the putative impact of known HY-presenting HLA alleles on future pregnancy outcome in women with at least three consecutive miscarriages following a birth [secondary recurrent miscarriage (SRM)]. HLA-A, -B, -DRB1, DRB3-5 and DQB1 genotyping was performed in 358 SRM patients and in 203 of their children born prior to the miscarriages. The subsequent live birth in women with boys prior to the miscarriages compared with girls is lower in women with HY-restricting HLA class II alleles [odds ratio (OR): 0.17 (0.1-0.4), P = 0.0001]. One HY-restricting HLA class II allele in women with firstborn boys significantly reduces the chances of a live birth [OR: 0.46 (0.2-0.9), P = 0.02]. Two HY-restricting HLA class II alleles further reduced this chance [OR: 0.21 (0.1-0.7), P = 0.02]. HY-restricting HLA class II did not reduce the chances of a live birth in SRM women with firstborn girls. HY-restricting HLA class II alleles are associated with a decreased chance of a live birth in SRM women with firstborn boys. These findings strongly indicate an aberrant maternal immune reaction against fetal HY-antigens in SRM. The results may shed light on the as-yet unknown immunological causes of SRM and may help understand the successful maternal acceptance of the fetal semi-allograft.


Subject(s)
Abortion, Habitual/genetics , H-Y Antigen/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility, Maternal-Fetal , Pregnancy/immunology , Abortion, Habitual/immunology , Female , H-Y Antigen/immunology , Histocompatibility Antigens Class II/immunology , Humans , Live Birth , Male , Pregnancy Outcome , Sex Characteristics
16.
Blood ; 112(1): 120-30, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18323414

ABSTRACT

During thymocyte development, the T-cell receptor (TCR) can discriminate major histocompatibility complex (MHC)/peptide ligands over a narrow range of affinities and translate subtle differences into functional fate decisions. How small differences in TCR input are translated into absolute differences in functional output is unclear. We examined the effects of galectin-1 ablation in the context of class-I-restricted thymocyte development. Galectin-1 expression opposed TCR partial agonist-driven positive selection, but promoted TCR agonist-driven negative selection of conventional CD8(+) T cells. Galectin-1 expression also promoted TCR agonist-driven CD8alphaalpha intestinal intraepithelial lymphocytes (IEL) development. Recombinant galectin-1 enhanced TCR binding to agonist/MHC complexes and promoted a negative-selection-signaling signature, reflected in intensified rapid and transient extracellular signal-regulated kinase (ERK) activation. In contrast, galectin-1 expression antagonized ERK activity in thymocytes undergoing positive selection. We propose that galectin-1 aids in discriminating TCR-directed fate decisions by promoting TCR binding to agonist/MHC complexes and enforcing agonist-driven signals, while opposing partial-agonist signals. In this way, galectin-1 widens the distinction between TCR-directed functional fate cues.


Subject(s)
Galectin 1/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Base Sequence , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , DNA Primers/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Galectin 1/genetics , Galectin 1/metabolism , Galectin 1/pharmacology , Gene Expression , H-Y Antigen/genetics , H-Y Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology
17.
Eur J Immunol ; 38(2): 391-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18203137

ABSTRACT

A developmental block is imposed on CD25(+)CD44(-) thymocytes at the beta-selection checkpoint in the absence of the pre T cell receptor (preTCR) alpha-chain, pTalpha. Early surface expression of a transgenic alphabeta TCR has been shown to partially circumvent this block, such that thymocytes progress to the CD4(+)CD8(+) double-positive stage. We wanted to analyze whether a restricting MHC element is required for alphabeta TCR-expressing double-negative (DN) thymocytes to overcome the developmental block in pTalpha-deficient animals. We used the HY-I knock-in model that endows thymocytes with alphabeta TCR expression in the DN compartment but has the advantage of physiological expression levels, in contrast to conventional TCR transgenes. On a pTalpha-deficient background, this HY-I TCR transgene 'rescued' CD25(+)CD44(-) thymocytes from apoptosis and enabled progression to later differentiation stages. On a non-selecting MHC background, however, pTalpha-deficient HY-I mice presented a pronounced reduction in numbers of splenocytes and thymocytes when compared to animals of selecting MHC genotype, showing that MHC restriction is necessary to drive HY-TCR-mediated rescue of pTalpha-deficient thymocytes.


Subject(s)
H-2 Antigens/immunology , Membrane Glycoproteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Female , H-2 Antigens/genetics , H-Y Antigen/genetics , H-Y Antigen/immunology , Histocompatibility Antigen H-2D , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Models, Immunological , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction/genetics , T-Lymphocytes/metabolism
18.
J Immunol ; 179(3): 1768-75, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641043

ABSTRACT

Adaptors play a critical role in regulating signaling pathways that control lymphocyte development and activation. Adaptor in lymphocytes of unknown function X (ALX) and Rlk/Itk-binding protein (RIBP) are adaptors related by structure and sequence, coexpressed in T cells. Mice deficient for each adaptor demonstrated that ALX and RIBP, respectively, negatively and positively regulate T cell activation in response to TCR/CD28 stimulation. However, these results did not preclude that they may function redundantly in other cell populations, or in response to other stimuli. Therefore, to understand the relationship between these related adaptors, ALX/RIBP-deficient mice were generated. We demonstrate that although ALX and RIBP are expressed throughout T cell development, T cell development occurs normally in these mice. Using the H-Y TCR transgenic model, positive and negative selection were found to proceed unimpeded in the absence of ALX and RIBP. We demonstrate that RIBP is also expressed in B cells; however, RIBP- and ALX/RIBP-deficient mice had normal B cell development, and responded equivalently to wild type in response to IgM, CD40, B cell-activating factor/B lymphocyte stimulator, CpG, and LPS. Interestingly, T cells deficient in both ALX and RIBP behaved similarly to those deficient in ALX alone during T cell activation in response to TCR/CD28, exhibiting increased IL-2 production, CD25 expression, and proliferation, thus showing that ALX deficiency masked the effect of RIBP deficiency. ALX/RIBP-deficient T cells did not have any alterations in either activation-induced cell death or Th1/2 polarization. Therefore, we did not find any functional redundancy or synergy during lymphocyte development, selection, activation, or survival in ALX/RIBP-deficient mice, demonstrating that these molecules function independently.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , H-Y Antigen/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Exp Biol Med (Maywood) ; 232(6): 780-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17526770

ABSTRACT

This study examines thymic nurse cell (TNC) function during T-cell development. It has been suggested that TNCs function in the removal of nonfunctional and/or apoptotic thymocytes and do not participate in major histocompatibility complex restriction. We analyzed TNCs isolated from both normal C57BL/6 mice and C57BL/6 TgN (TCRHY) mice (HY-TCR transgenic mice). Using confocal microscopic analyses of TNCs isolated from C57BL/6 animals, we showed that 75%-78% of the enclosed thymocyte subset was viable, and 87%-90% of these cells expressed both CD4 and CD8. CD4 and CD8 also were expressed on TNC thymocytes isolated from both male and female HY-TCR transgenic mice. The transgenic female thymus was shown to have 17 times more TNCs per milligram of thymus than the transgenic male thymus. TNCs from HY-TCR transgenic females were 8-10 microm larger than transgenic male TNCs, and the female TNCs contained five times more thymocytes within intracytoplasmic vacuoles, with less than 4% apoptosis. However, more than 42% of the thymocytes within transgenic male TNCs were apoptotic. The large number and size of TNCs containing viable thymocytes in the female transgenic thymus suggest that TNC function is not limited to the removal of apoptotic thymocytes. We believe that the selective uptake of viable double-positive thymocytes by TNCs in C57BL/6 and HY-TCR transgenic female mice provides evidence that this interaction occurs during the process of major histocompatibility complex restriction.


Subject(s)
Mice, Transgenic/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Female , H-Y Antigen/genetics , In Situ Nick-End Labeling , Major Histocompatibility Complex , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , Self Tolerance , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
20.
PLoS One ; 1: e42, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17183671

ABSTRACT

BACKGROUND: Minor Histocompatibility (H) antigen mismatches significantly influence the outcome of HLA-matched allogeneic stem cell transplantation. The molecular identification of human H antigens is increasing rapidly. In parallel, clinical application of minor H antigen typing has gained interest. So far, relevant and simple tools to analyze the minor H antigens in a quick and reliable way are lacking. METHODOLOGY AND FINDINGS: We developed a uniform PCR with sequence-specific primers (PCR-SSP) for 10 different autosomal minor H antigens and H-Y. This genomic minor H antigen typing methodology allows easy incorporation in the routine HLA typing procedures. DNA from previously typed EBV-LCL was used to validate the methodology. To facilitate easy interpretation for clinical purposes, a minor H database named dbMinor (http://www.lumc.nl/dbminor) was developed. Input of the minor H antigen typing results subsequently provides all relevant information for a given patient/donor pair and additional information on the putative graft-versus-host, graft-versus-tumor and host-versus-graft reactivities. SIGNIFICANCE: A simple, uniform and rapid methodology was developed enabling determination of minor H antigen genotypes of all currently identified minor H antigens. A dbMinor database was developed to interpret the genomic typing for its potential clinical relevance. The combination of the minor H antigen genomic typing methodology with the online dbMinor database and applications facilitates the clinical application of minor H antigens anti-tumor targets after stem cell transplantation.


Subject(s)
Histocompatibility Testing/methods , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Loci , Alleles , Base Sequence , DNA Primers/genetics , Databases, Genetic , H-Y Antigen/genetics , HLA Antigens/genetics , Humans , Polymerase Chain Reaction/methods , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...