Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 9: 3038, 2018.
Article in English | MEDLINE | ID: mdl-30728823

ABSTRACT

Different profiles of alloantibody responses are observed in the clinic, with those that persist, often despite targeted treatment, associated with poorer long-term transplant outcomes. Although such responses would suggest an underlying germinal center (GC) response, the relationship to cellular events within the allospecific B cell population is unclear. Here we examine the contribution of germinal center (GC) humoral alloimmunity to chronic antibody mediated rejection (AMR). A murine model of chronic AMR was developed in which T cell deficient (Tcrbd-/-) C57BL/6 recipients were challenged with MHC-mismatched BALB/c heart allografts and T cell help provided by reconstituting with 103 "TCR75" CD4 T cells that recognize self-restricted allopeptide derived from the H-2Kd MHC class I alloantigen. Reconstituted recipients developed Ig-switched anti-Kd alloantibody responses that were slow to develop, but long-lived, with confocal immunofluorescence and flow cytometric characterization of responding H-2Kd-allospecific B cells confirming persistent splenic GC activity. This was associated with T follicular helper (TFH) cell differentiation of the transferred TCR75 CD4 T cells. Heart grafts developed progressive allograft vasculopathy, and were rejected chronically (MST 50 days), with explanted allografts displaying features of humoral vascular rejection. Critically, late alloantibody responses were abolished, and heart grafts survived indefinitely, in recipients reconstituted with Sh2d1a-/- TCR75 CD4 T cells that were genetically incapable of providing TFH cell function. The GC response was associated with affinity maturation of the anti-Kd alloantibody response, and its contribution to progression of allograft vasculopathy related principally to secretion of alloantibody, rather than to enhanced alloreactive T cell priming, because grafts survived long-term when B cells could present alloantigen, but not secrete alloantibody. Similarly, sera sampled at late time points from chronically-rejecting recipients induced more vigorous donor endothelial responses in vitro than sera sampled earlier after transplantation. In summary, our results suggest that chronic AMR and progression of allograft vasculopathy is dependent upon allospecific GC activity, with critical help provided by TFH cells. Clinical strategies that target the TFH cell subset may hold therapeutic potential. This work is composed of two parts, of which this is Part II. Please read also Part I: Alsughayyir et al., 2019.


Subject(s)
Germinal Center/immunology , Graft Rejection/immunology , Heart Transplantation/adverse effects , Immunity, Humoral , Isoantibodies/immunology , Allografts/immunology , Animals , Chronic Disease , Disease Models, Animal , Disease Progression , Germinal Center/metabolism , Humans , Isoantibodies/metabolism , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocardium/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Transplantation, Homologous/adverse effects
2.
Front Immunol ; 9: 3039, 2018.
Article in English | MEDLINE | ID: mdl-30740108

ABSTRACT

Humoral alloimmunity is now recognized as a major determinant of transplant outcome. MHC glycoprotein is considered a typical T-dependent antigen, but the nature of the T cell alloresponse that underpins alloantibody generation remains poorly understood. Here, we examine how the relative frequencies of alloantigen-specific B cells and helper CD4 T cells influence the humoral alloimmune response and how this relates to antibody-mediated rejection (AMR). An MHC-mismatched murine model of cardiac AMR was developed, in which T cell help for alloantibody responses in T cell deficient (Tcrbd-/-) C57BL/6 recipients against donor H-2Kd MHC class I alloantigen was provided by adoptively transferred "TCR75" CD4 T cells that recognize processed H-2Kd allopeptide via the indirect-pathway. Transfer of large numbers (5 × 105) of TCR75 CD4 T cells was associated with rapid development of robust class-switched anti-H-2Kd humoral alloimmunity and BALB/c heart grafts were rejected promptly (MST 9 days). Grafts were not rejected in T and B cell deficient Rag2-/- recipients that were reconstituted with TCR75 CD4 T cells or in control (non-reconstituted) Tcrbd-/- recipients, suggesting that the transferred TCR75 CD4 T cells were mediating graft rejection principally by providing help for effector alloantibody responses. In support, acutely rejecting BALB/c heart grafts exhibited hallmark features of acute AMR, with widespread complement C4d deposition, whereas cellular rejection was not evident. In addition, passive transfer of immune serum from rejecting mice to Rag2-/- recipients resulted in eventual BALB/c heart allograft rejection (MST 20 days). Despite being long-lived, the alloantibody responses observed at rejection of the BALB/c heart grafts were predominantly generated by extrafollicular foci: splenic germinal center (GC) activity had not yet developed; IgG secreting cells were confined to the splenic red pulp and bridging channels; and, most convincingly, rapid graft rejection still occurred when recipients were reconstituted with similar numbers of Sh2d1a-/- TCR75 CD4 T cells that are genetically incapable of providing T follicular helper cell function for generating GC alloimmunity. Similarly, alloantibody responses generated in Tcrbd-/- recipients reconstituted with smaller number of wild-type TCR75 CD4 T cells (103), although long-lasting, did not have a discernible extrafollicular component, and grafts were rejected much more slowly (MST 50 days). By modeling antibody responses to Hen Egg Lysozyme protein, we confirm that a high ratio of antigen-specific helper T cells to B cells favors development of the extrafollicular response, whereas GC activity is favored by a relatively high ratio of B cells. In summary, a relative abundance of helper CD4 T cells favors development of strong extrafollicular alloantibody responses that mediate acute humoral rejection, without requirement for GC activity. This work is composed of two parts, of which this is Part I. Please read also Part II: Chhabra et al., 2019.


Subject(s)
B-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/adverse effects , Isoantibodies/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Allografts/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Disease Progression , Graft Rejection/blood , Humans , Isoantibodies/metabolism , Isoantigens/immunology , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocardium/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Transplantation, Homologous/adverse effects
3.
J Heart Lung Transplant ; 29(4): 417-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20188594

ABSTRACT

BACKGROUND: Coronary artery vasculopathy (CAV) is the major life-limiting factor in cardiac transplantation, after 1 year. Antibody-mediated rejection (AMR) has been associated with development of both acute and chronic rejection. We analyzed endomyocardial biopsies for pathologic markers of AMR (C4d and C3d), from the first 2 years post-transplantation, to determine complement deposition in relation to the development of CAV. METHODS: A retrospective, matched-pair study was used. Group 1 subjects (n = 26) were CAV-negative at 8 years, and Group 2 (n = 26) had angiographically detectable CAV at 4 years. Biopsies from six time-points were studied (total = 282). Immunohistochemistry was performed for C4d, C3d and CD68. Biopsies were graded for rejection using ISHLT criteria. RESULTS: Although CAV was not significantly associated with C4d deposition, it was associated with C3d deposition (p = 0.043). Only 4% of C4d and 5% of C3d biopsies were completely negative. Group 1 had 6 AMR-positive biopsies, with Group 2 having 8. There was no significant relationship between acute cellular rejection or AMR events and CAV. CONCLUSIONS: This study demonstrates that complement deposition is a frequent occurrence in the first 2 years post-transplantation. Although acute rejection is a known risk factor for CAV, in this study the relationship was found not to be significant. No relationship was found with the development of CAV and histologic features of AMR, when assessed by C4d deposition alone. However, an association between C3d deposition and the development of CAV was determined in this study group, suggesting that complement activation may play a role in the pathogenesis of CAV.


Subject(s)
Complement C3d/metabolism , Complement C4b/metabolism , Coronary Artery Disease/physiopathology , Heart Transplantation/physiology , Peptide Fragments/metabolism , Adult , Antibodies/physiology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Biopsy , Coronary Artery Disease/epidemiology , Coronary Artery Disease/immunology , Female , Graft Rejection/complications , Graft Rejection/immunology , Graft Rejection/physiopathology , Heart Transplantation/immunology , Humans , Male , Matched-Pair Analysis , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...