6 research outputs found

    Cytotoxicity and T-B Cell Crosstalk in Belatacept-Treated Kidney Transplant Patients

    Get PDF
    In this dissertation, we aimed to learn more about the immune mechanisms involved in alloreactivity in patients treated with belatacept or tacrolimus after kidney transplantation. In particular, we sought to explain the higher acute rejection rate in belatacept-treated patients by studying cytotoxic cell populations which are potentially less susceptible to co-stimulatory inhibition, and to explain the lower incidence of DSA in previous trials by studying the effects of belatacept on Tfh-B cell interaction. We found that next to CD28- T cells also a part of CD28+ T cells are not susceptible to inhibition by belatacept. These cells could still divide and produce cytokines in the presence of belatacept. In addition, belatacept was not as effective as tacrolimus in preventing Tfh-B cell interaction and the subsequent formation of plasmablasts. Additionally, we tried to characterize belatacept-resistant rejection clinically and immunologically, and to find a biomarker to distinguish between patients who will reject under belatacept-treatment and those who will not. We found that belatacept-treated patients had a higher acute rejection risk compared to tacrolimus-treated patients (55% versus 10%), and that these rejections could not be predicted by cellular biomarkers before transplantation, i.e., CD8+CD28- T cells, CD4+CD57+PD-1- T cells and CD8+CD28++ EMRA T cells. Baseline characteristics did not differ between (future) rejectors and non-rejectors in belatacept-treated patients. Mostly effector-memory T cells, both CD28+ and CD28-, infiltrated a rejected kidney graft during belatacept-treatment. These cells produced large amounts of IFN-gamma and granzyme B. For now, belatacept does not meet the requirements to be the new corner stone in immunosuppression after kidney transplantation, because short-term outcomes are inferior to tacrolimus. However, belatacept-treatment could be beneficial and increase quality of life for a selected group of patients. The search for an immunosuppressive drug or combination of immunosuppressants, possibly including belatacept, to further improve kidney transplant patients’ outcomes continues

    Down-regulation of surface CD28 under belatacept treatment: An escape mechanism for antigen-reactive T-cells

    Get PDF
    Background The co-stimulatory inhibitor of the CD28-CD80/86-pathway, belatacept, allows calcineurininhibitor-free immunosuppression in kidney transplantation. However, aggressive T-cell mediated allogeneic responses have been observed in belatacept-treated patients, which could be explained by effector-memory T-cells that lack membrane expression of CD28, i.e. CD28-negative (CD28NULL) T-cells. CD28-positive (CD28POS) T-cells that down regulate their surface CD28 after allogeneic stimulation could also pose a threat against the renal graft. The aim of this study was to investigate this potential escape mechanism for CD28POS T-cells under belatacept treatment. Materials & Methods PBMCs, isolated T-cell memory subsets and isolated CD28POS T-cells were obtained from end-stage renal disease (ESRD) patients and co-cultured with allo-antigen in the presence of belatacept to mimic allogeneic reactions in kidney-transplant patients under belatacept treatment. As a control, IgG was used in the absence of belatacept. Results Despite high in vitro belatacept concentrations, a residual T-cell growth of ±30% was observed compared to the IgG control after allogeneic stimulation. Of the alloreactive Tcells, the majority expressed an effector-memory phenotype. This predominance for effector-memory T-cells within the proliferated cells was even larger when a higher dose of belatacept was added. Contrary to isolated naïve and central-memory T cells, isolated effectormemory T cells could not be inhibited by belatacept in differentiation or allogeneic IFNγ production. The proportion of CD28-positive T cells was lower within the proliferated T cell population, but was still substantial. A fair number of the isolated initially CD28POS T-cells differentiated into CD28NULL T-cells, which made them not targetable by belatacept. These induced CD28NULL T-cells were not anergic as they produced high amounts of IFNγ upon allogeneic stimulat

    Improved Glucose Tolerance in a Kidney Transplant Recipient With Type 2 Diabetes Mellitus After Switching From Tacrolimus To Belatacept: A Case Report and Review of Potential Mechanisms

    Get PDF
    The introduction of immunosuppressant belatacept, an inhibitor of the CD28-80/86 pathway, has improved 1-year outcomes in kidney transplant recipients with preexistent diabetes mellitus and has also reduced the risk of posttransplant diabetes mellitus. So far, no studies have compared a tacrolimus-based with a belatacept-based immunosuppressive regimen with regard to improving glucose tolerance after kidney transplantation. Here, we present the case of a 54-year-old man with type 2 diabetes mellitus who was converted from belatacept to tacrolimus 1 year after a successful kidney transplantation. Thereafter, he quickly developed severe hyperglycemia, and administration of insulin was needed to improve metabolic control. Six months after this episode, he was converted back to belatacept because of nausea, diarrhea, and hyperglycemia. After switching back to belatacept and within 4 days aft

    Pharmacodynamic Monitoring of Tacrolimus-based Immunosuppression in CD14+ Monocytes after Kidney Transplantation

    Get PDF
    Background: Monocytes significantly contribute to ischemia-reperfusion injury and allograft rejection after kidney transplantation. However, the knowledge about the effects of immunosuppressive drugs on monocyte activation is limited. Conventional pharmacokinetic methods for immunosuppressive drug monitoring are not cell type–specific. In this study, phosphorylation of 3 signaling proteins was measured to determine the pharmacodynamic effects of immunosuppression on monocyte activation in kidney transplant patients. Methods: Blood samples from 20 kidney transplant recipients were monitored before and during the first year after transplantation. All patients received induction therapy with basiliximab, followed by tacrolimus (TAC), mycophenolate mofetil, and prednisolone maintenance therapy. TAC whole-blood predose concentrations were determined using an antibody-conjugated magnetic immunoassay. Samples were stimulated with phorbol 12-myristate 13-acetate (PMA)/ionomycin, and phosphorylation of p38MAPK, ERK, and Akt in CD14+ monocytes was quantified by phospho-specific flow cytometry. Results: Phosphorylation of p38MAPK and Akt in monocytes of immunosuppressed recipients was lower after 360 days compared with before transplantation in the unstimulated samples [mean reduction in median fluorescence intensity 36%; range −28% to 77% for p-p38MAPK and 20%; range −22% to 53% for p-Akt; P < 0.05]. P-ERK was only decreased at day 4 after transplantation (mean inhibition 23%; range −52% to 73%; P < 0.05). At day 4, when the highest whole-blood predose TAC concentrations were measured, p-p38MAPK and p-Akt, but not p-ERK, correlated inversely with TAC (rs = −0.65; P = 0.01 and rs = −0.58; P = 0.03, respectively). Conclusions: Immunosuppressive drug combination therapy partially inhibits monocyte activation pathways after kidney transplantation. This inhibition can be determined by phospho-specific flow cytometry, which enables the assessment of the pharmacodynamic effects of immunosuppressive drugs in a cell type–specific manner

    Follicular T helper cells and humoral reactivity in kidney transplant patients

    No full text
    Summary: Memory B cells play a pivotal role in alloreactivity in kidney transplantation. Follicular T helper (Tfh) cells play an important role in the differentiation of B cells into immunoglobulin-producing plasmablasts [through interleukin (IL)-21]. It is unclear to what extent this T cell subset regulates humoral alloreactivity in kidney transplant patients, therefore we investigated the absolute numbers and function of peripheral Tfh cells (CD4POSCXCR5POS T cells) in patients before and after transplantation. In addition, we studied their relationship with the presence of donor-specific anti-human leucocyte antigen (HLA) antibodies (DSA), and the presence of Tfh cells in rejection biopsies. After transplantation peripheral Tfh cell numbers remained stable
    corecore