Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35674134

ABSTRACT

Dominant infectious tolerance explains how brief tolerance-inducing therapies result in lifelong tolerance to donor antigens and "linked" third-party antigens, while recipient sensitization and ensuing immunological memory prevent the successful induction of transplant tolerance. In this study, we juxtapose these 2 concepts to test whether mechanisms of dominant infectious tolerance can control a limited repertoire of memory T and B cells. We show that sensitization to a single donor antigen is sufficient to prevent stable transplant tolerance, rendering it unstable. Mechanistic studies revealed that recall antibody responses and memory CD8+ T cell expansion were initially controlled, but memory CD4+Foxp3- T cell (Tconv) responses were not. Remarkably, naive donor-specific Tconvs at tolerance induction also acquired a resistance to tolerance, proliferating and acquiring a phenotype similar to memory Tconvs. This phenomenon of "linked sensitization" underscores the challenges of reprogramming a primed immune response toward tolerance and identifies a potential therapeutic checkpoint for synergizing with costimulation blockade to achieve transplant tolerance in the clinic.


Subject(s)
CD4-Positive T-Lymphocytes , Transplantation Tolerance , Animals , Graft Rejection/prevention & control , Immune Tolerance , Mice , Mice, Inbred C57BL
2.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33393512

ABSTRACT

Immunological tolerance to semiallogeneic fetuses is necessary to achieving successful first pregnancy and permitting subsequent pregnancies with the same father. Paradoxically, pregnancy is an important cause of sensitization, resulting in the accelerated rejection of offspring-matched allografts. The underlying basis for divergent outcomes following reencounter of the same alloantigens on transplanted organs versus fetuses in postpartum females is incompletely understood. Using a mouse model that allows concurrent tracking of endogenous fetus-specific T and B cell responses in a single recipient, we show that semiallogeneic pregnancies simultaneously induce fetus-specific T cell tolerance and humoral sensitization. Pregnancy-induced antibodies, but not B cells, impeded transplantation tolerance elicited by costimulation blockade to offspring-matched cardiac grafts. Remarkably, in B cell-deficient mice, allogeneic pregnancy enabled the spontaneous acceptance of fetus-matched allografts. The presence of pregnancy-sensitized B cells that cannot secrete antibodies at the time of heart transplantation was sufficient to precipitate rejection and override pregnancy-established T cell tolerance. Thus, while induction of memory B cells and alloantibodies by pregnancies establishes formidable barriers to transplant success for multigravid women, our observations raise the possibility that humoral desensitization will not only improve transplantation outcomes, but also reveal an unexpected propensity of multiparous recipients to achieve tolerance to offspring-matched allografts.


Subject(s)
B-Lymphocytes/immunology , Fetal Tissue Transplantation , Fetus/immunology , Isoantibodies/immunology , T-Lymphocytes/immunology , Transplantation Tolerance , Allografts , Animals , Female , Mice , Mice, Transgenic , Pregnancy
3.
J Clin Invest ; 130(7): 3453-3466, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32452834

ABSTRACT

The absence of alloantibodies is a feature of transplantation tolerance. Although the lack of T cell help has been evoked to explain this absence, herein we provide evidence for B cell-intrinsic tolerance mechanisms. Using a murine model of heart tolerance, we showed that alloreactive B cells were not deleted but rapidly lost their ability to differentiate into germinal center B cells and secrete donor-specific antibodies. We inferred that tolerant alloreactive B cells retained their ability to sense alloantigen because they continued to drive T cell maturation into CXCR5+PD-1+ T follicular helper cells. Unexpectedly, dysfunctional alloreactive B cells acquired the ability to inhibit antibody production by new naive B cells in an antigen-specific manner. Thus, tolerant alloreactive B cells contribute to transplantation tolerance by foregoing germinal center responses while retaining their ability to function as antigen-presenting cells and by actively suppressing de novo alloreactive B cell responses.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Isoantibodies/immunology , Isoantigens/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transplantation Tolerance , Animals , B-Lymphocytes/pathology , Female , Germinal Center/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/pathology
4.
Am J Transplant ; 20(10): 2675-2685, 2020 10.
Article in English | MEDLINE | ID: mdl-32243663

ABSTRACT

Active antibody-mediated rejection (AMR) is a potentially devastating complication and consistently effective treatment remains elusive. We hypothesized that the reversal of acute AMR requires rapid elimination of antibody-secreting plasma cells (PC) with a proteasome inhibitor, bortezomib, followed by the sustained inhibition of PC generation with CTLA4-Ig or belatacept (B/B). We show in mice that B/B therapy selectively depleted mature PC producing donor-specific antibodies (DSA) and reduced DSA, when administered after primary and secondary DSA responses had been established. A pilot investigation was initiated to treat six consecutive patients with active AMR with B/B. Compassionate use of this regimen was initiated for the first patient who developed early, severe acute AMR that did not respond to steroids, plasmapheresis, and intravenous immunoglobulin after his third kidney transplant. B/B treatment resulted in a rapid reversal of AMR, leading us to treat five additional patients who also resolved their acute AMR episode and had sustained disappearance of circulating DSA for ≤30 months. This study provides a proof-of-principle demonstration that mouse models can identify mechanistically rational therapies for the clinic. Follow-up investigations with a more stringent clinical design are warranted to test whether B/B improves on the standard of care for the treatment of acute AMR.


Subject(s)
Kidney Transplantation , Abatacept/therapeutic use , Animals , Antibody Formation , Bortezomib/therapeutic use , Graft Rejection/drug therapy , Graft Rejection/prevention & control , Humans , Isoantibodies , Mice
5.
Front Immunol ; 9: 1385, 2018.
Article in English | MEDLINE | ID: mdl-29973932

ABSTRACT

Despite numerous advances in the definition of a role for regulatory T cells (Tregs) in facilitating experimental transplantation tolerance, and ongoing clinical trials for Treg-based therapies, critical issues related to the optimum dosage, antigen-specificity, and Treg-friendly adjunct immunosuppressants remain incompletely resolved. In this study, we used a tractable approach of MHC tetramers and flow cytometry to define the fate of conventional (Tconvs) and Tregs CD4+ T cells that recognize donor 2W antigens presented by I-Ab on donor and recipient antigen-presenting cells (APCs) in a mouse cardiac allograft transplant model. Our study shows that these endogenous, donor-reactive Tregs comparably accumulate in the spleens of recipients undergoing acute rejection or exhibiting costimulation blockade-induced tolerance. Importantly, this expansion was not detected when analyzing bulk splenic Tregs. Systemically, the distinguishing feature between tolerance and rejection was the inhibition of donor-reactive conventional T cell (Tconv) expansion in tolerance, translating into increased percentages of splenic FoxP3+ Tregs within the 2W:I-Ab CD4+ T cell subset compared to rejection (~35 vs. <5% in tolerance vs. rejection). We further observed that continuous administration of rapamycin, cyclosporine A, or CTLA4-Ig did not facilitate donor-specific Treg expansion, while all three drugs inhibited Tconv expansion. Finally, donor-specific Tregs accumulated comparably in rejecting tolerant allografts, whereas tolerant grafts harbored <10% of the donor-specific Tconv numbers observed in rejecting allografts. Thus, ~80% of 2W:I-Ab CD4+ T cells in tolerant allografts expressed FoxP3+ compared to ≤10% in rejecting allografts. A similar, albeit lesser, enrichment was observed with bulk graft-infiltrating CD4+ cells, where ~30% were FoxP3+ in tolerant allografts, compared to ≤10% in rejecting allografts. Finally, we assessed that the phenotype of 2W:I-Ab Tregs and observed that the percentages of cells expressing neuropilin-1 and CD73 were significantly higher in tolerance compared to rejection, suggesting that these Tregs may be functionally distinct. Collectively, the analysis of donor-reactive, but not of bulk, Tconvs and Tregs reveal a systemic signature of tolerance that is stable and congruent with the signature within tolerant allografts. Our data also underscore the importance of limiting Tconv expansion for high donor-specific Tregs:Tconv ratios to be successfully attained in transplantation tolerance.

6.
Front Immunol ; 8: 1169, 2017.
Article in English | MEDLINE | ID: mdl-28970838

ABSTRACT

Clinical observations that kidney transplant recipients receiving belatacept who experienced T cell-mediated acute rejection can be successfully treated and subsequently maintained on belatacept-based immunosuppression suggest that belatacept is able to control memory T cells. We recently reported that treatment with CTLA4-Ig from day 6 posttransplantation successfully rescues allografts from acute rejection in a BALB/c to C57BL/6 heart transplant model, in part, by abolishing B cell germinal centers and reducing alloantibody titers. Here, we show that CTLA4-Ig is additionally able to inhibit established T cell responses independently of B cells. CTLA4-Ig inhibited the in vivo cytolytic activity of donor-specific CD8+ T cells, and the production of IFNγ by graft-infiltrating T cells. Delayed CTLA4-Ig treatment did not reduce the numbers of graft-infiltrating T cells nor prevented the accumulation of antigen-experienced donor-specific memory T cells in the spleen. Nevertheless, delayed CTLA4-Ig treatment successfully maintained long-term graft acceptance in the majority of recipients that had experienced a rejection crisis, and enabled the acceptance of secondary BALB/c heart grafts transplanted 30 days after the first transplantation. In summary, we conclude that delayed CTLA4-Ig treatment is able to partially halt ongoing T cell-mediated acute rejection. These findings extend the functional efficacy of CTLA4-Ig therapy to effector T cells and provide an explanation for why CTLA4-Ig-based immunosuppression in the clinic successfully maintains long-term graft survival after T cell-mediated rejection.

7.
Transplantation ; 101(11): 2671-2681, 2017 11.
Article in English | MEDLINE | ID: mdl-28604446

ABSTRACT

Whether a transplanted allograft is stably accepted, rejected, or achieves immunological tolerance is dependent on the frequency and function of alloreactive lymphocytes, making the identification and analysis of alloreactive T and B cells in transplant recipients critical for understanding mechanisms, and the prediction of allograft outcome. In animal models, tracking the fate of graft-reactive T and B cells allows investigators to uncover their biology and develop new therapeutic strategies to protect the graft. In the clinic, identification and quantification of graft-reactive T and B cells allows for the early diagnosis of immune reactivity and therapeutic intervention to prevent graft loss. In addition to rejection, probing of T and B cell fate in vivo provides insights into the underlying mechanisms of alloimmunity or tolerance that may lead to biomarkers predicting graft fate. In this review, we discuss existing and developing approaches to track and analyze alloreactive T and B cells in mice and humans and provide examples of discoveries made utilizing these techniques. These approaches include mixed lymphocyte reactions, trans-vivo delayed-type hypersensitivity, enzyme-linked immunospot assays, the use of antigen receptor transgenic lymphocytes, and utilization of peptide-major histocompatibility multimers, along with imaging techniques for static multiparameter analysis or dynamic in vivo tracking. Such approaches have already refined our understanding of the alloimmune response and are pointing to new ways to improve allograft outcomes in the clinic.


Subject(s)
B-Lymphocytes/immunology , Graft Rejection/prevention & control , Graft Survival , Immunologic Techniques , Lymphocyte Activation , Organ Transplantation , T-Lymphocytes/immunology , Transplantation Tolerance , Allografts , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Graft Rejection/blood , Graft Rejection/immunology , Graft Survival/drug effects , Histocompatibility , Humans , Immunosuppressive Agents/therapeutic use , Isoantibodies/blood , Lymphocyte Activation/drug effects , Mice , Models, Animal , Organ Transplantation/adverse effects , Plasma Cells/immunology , Risk Factors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation Tolerance/drug effects , Treatment Outcome
8.
JCI Insight ; 2(9)2017 May 04.
Article in English | MEDLINE | ID: mdl-28469082

ABSTRACT

Despite recent evidence of improved graft outcomes and safety, the high incidence of early acute cellular rejection with belatacept, a high-affinity CTLA4-Ig, has limited its use in clinical transplantation. Here we define how the incomplete control of endogenous donor-reactive memory T cells results in belatacept-resistant rejection in an experimental model of BALB/c.2W-OVA donor heart transplantation into C57BL/6 recipients presensitized to donor splenocytes. These sensitized mice harbored modestly elevated numbers of endogenous donor-specific memory T cells and alloantibodies compared with naive recipients. Continuous CTLA4-Ig treatment was unexpectedly efficacious at inhibiting endogenous graft-reactive T cell expansion but was unable to inhibit late CD4+ and CD8+ T cell infiltration into the allografts, and rejection was observed in 50% of recipients by day 35 after transplantation. When CTLA4-Ig was combined with the sphingosine 1-phosphate receptor-1 (S1PR1) functional antagonist FTY720, alloantibody production was inhibited and donor-specific IFN-γ-producing T cells were reduced to levels approaching nonsensitized tolerant recipients. Late T cell recruitment into the graft was also restrained, and graft survival improved with this combination therapy. These observations suggest that a rational strategy consisting of inhibiting memory T cell expansion and trafficking into the allograft with CTLA4-Ig and FTY720 can promote allograft survival in allosensitized recipients.

9.
Transplantation ; 100(8): 1683-91, 2016 08.
Article in English | MEDLINE | ID: mdl-27362308

ABSTRACT

BACKGROUND: The dual role of B cells as drivers and suppressors of the immune responses have underscored the need to trace the fate of B cells recognizing donor major histocompatibility complex class I and class II after allograft transplantation. METHODS: In this study, we used donor class II tetramers to trace the fate of I-E-specific B cells after immunization with BALB/c spleen cells or cardiac transplantation, in naive or sensitized C57BL/6 recipients. We combined this approach with genetic lineage tracing of memory B cells in activation-induced cytidine deaminase regulated Cre transgenic mice crossed to the ROSA26-enhanced yellow fluorescent protein reporter mice to track endogenous I-E-specific memory B cell generation. RESULTS: Immunization with BALB/c splenocytes or heart transplantation induced an expansion and differentiation of I-E-specific B cells into germinal center B cells, whereas BALB/c heart transplantation into sensitized recipients induced the preferential differentiation into antibody-secreting cells. A 10.8-fold increase in the frequency of I-E-specific memory B cells was observed by day 42 postimmunization. Treatment with CTLA4-Ig starting on day 0 or day 7 postimmunization abrogated I-E-specific memory B cell generation and sensitized humoral responses, but not if treatment commenced on day 14. CONCLUSIONS: The majority of donor-specific memory B cells are generated between days 7 and 14 postimmunization, thus revealing a flexible timeframe whereby delayed CTLA4-Ig administration can inhibit sensitization and the generation of memory graft-reactive B cells.


Subject(s)
Abatacept/administration & dosage , B-Lymphocytes/drug effects , Cell Lineage , Cell Proliferation/drug effects , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Histocompatibility Antigens Class II/immunology , Immunologic Memory , Immunosuppressive Agents/administration & dosage , Lymphocyte Activation/drug effects , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Cell Tracking/methods , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Disease Models, Animal , Drug Administration Schedule , Genotype , Graft Rejection/blood , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/drug effects , Histocompatibility Antigens Class II/blood , Integrases/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RNA, Untranslated/genetics , Time Factors
10.
J Immunol ; 191(1): 488-99, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23709681

ABSTRACT

Prevention of chronic graft-versus-host disease (cGVHD) remains a major challenge in allogeneic hematopoietic cell transplantation (HCT) owing to limited understanding of cGVHD pathogenesis and lack of appropriate animal models. In this study, we report that, in classical acute GVHD models with C57BL/6 donors and MHC-mismatched BALB/c recipients and with C3H.SW donors and MHC-matched C57BL/6 recipients, GVHD recipients surviving for >60 d after HCT developed cGVHD characterized by cutaneous fibrosis, tissue damage in the salivary gland, and the presence of serum autoantibodies. Donor CD8(+) T cells were more potent than CD4(+) T cells for inducing cGVHD. The recipient thymus and de novo-generated, donor-derived CD4(+) T cells were required for induction of cGVHD by donor CD8(+) T cells but not by donor CD4(+) T cells. Donor CD8(+) T cells preferentially damaged recipient medullary thymic epithelial cells and impaired negative selection, resulting in production of autoreactive CD4(+) T cells that perpetuated damage to the thymus and augmented the development of cGVHD. Short-term anti-CD4 mAb treatment early after HCT enabled recovery from thymic damage and prevented cGVHD. These results demonstrate that donor CD8(+) T cells cause cGVHD solely through thymic-dependent mechanisms, whereas CD4(+) T cells can cause cGVHD through either thymic-dependent or independent mechanisms.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chronic Disease , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Thymus Gland/transplantation
11.
J Immunol ; 189(1): 222-33, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649197

ABSTRACT

We reported that both donor CD4(+) T and B cells in transplants were required for induction of an autoimmune-like chronic graft-versus-host disease (cGVHD) in a murine model of DBA/2 donor to BALB/c recipient, but mechanisms whereby donor B cells augment cGVHD pathogenesis remain unknown. In this study, we report that, although donor B cells have little impact on acute GVHD severity, they play an important role in augmenting the persistence of tissue damage in the acute and chronic GVHD overlapping target organs (i.e., skin and lung); they also markedly augment damage in a prototypical cGVHD target organ, the salivary gland. During cGVHD pathogenesis, donor B cells are activated by donor CD4(+) T cells to upregulate MHC II and costimulatory molecules. Acting as efficient APCs, donor B cells augment donor CD4(+) T clonal expansion, autoreactivity, IL-7Rα expression, and survival. These qualitative changes markedly augment donor CD4(+) T cells' capacity in mediating autoimmune-like cGVHD, so that they mediate disease in the absence of donor B cells in secondary recipients. Therefore, a major mechanism whereby donor B cells augment cGVHD is through augmenting the clonal expansion, differentiation, and survival of pathogenic CD4(+) T cells.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/transplantation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Up-Regulation/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cell Survival/genetics , Cell Survival/immunology , Chronic Disease , Clone Cells , Disease Models, Animal , Graft vs Host Disease/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Salivary Glands/immunology , Salivary Glands/pathology , Skin/immunology , Skin/pathology , Up-Regulation/genetics
12.
J Immunol ; 186(5): 2739-49, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21263067

ABSTRACT

Foxp3(+) regulatory T (Treg) cells include thymic-derived natural Treg and conventional T-derived adaptive Treg cells. Both are proposed to play important roles in downregulating inflammatory immune responses. However, the mechanisms of Treg expansion in inflammatory environments remain unclear. In this study, we report that, in an autoimmune-like graft-versus-host disease model of DBA/2 (H-2(d)) donor to BALB/c (H-2(d)) recipients, donor Treg cells in the recipients predominantly originated from expansion of natural Treg cells and few originated from adaptive Treg cells. In vivo neutralization of IFN-γ resulted in a marked reduction of donor natural Treg expansion and exacerbation of graft-versus-host disease, which was associated with downregulation of host APC expression of B7H1. Furthermore, host APC expression of B7H1 was shown to augment donor Treg survival and expansion. Finally, donor Treg interactions with host APCs via B7.1/B7H1 but not PD-1/B7H1 were demonstrated to be critical in augmenting donor Treg survival and expansion. These studies have revealed a new immune regulation loop consisting of T cell-derived IFN-γ, B7H1 expression by APCs, and B7.1 expression by Treg cells.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Surface/physiology , Apoptosis Regulatory Proteins/physiology , B7-1 Antigen/physiology , Cell Differentiation/immunology , Membrane Glycoproteins/physiology , Peptides/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B7-1 Antigen/biosynthesis , B7-H1 Antigen , Cell Differentiation/genetics , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immunity, Innate/genetics , Inducible T-Cell Co-Stimulator Ligand , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peptides/antagonists & inhibitors , Programmed Cell Death 1 Receptor , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
13.
J Immunol ; 186(2): 856-68, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149609

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is considered an autoimmune-like disease mediated by donor CD4(+) T cells, but the origin of the autoreactive T cells is still controversial. In this article, we report that the transplantation of DBA/2 donor spleen cells into thymectomized MHC-matched allogeneic BALB/c recipients induced autoimmune-like cGVHD, although not in control syngeneic DBA/2 recipients. The donor-type CD4(+) T cells from the former but not the latter recipients induced autoimmune-like manifestations in secondary allogeneic BALB/c as well as syngeneic DBA/2 recipients. Transfer of donor-type CD4(+) T cells from secondary DBA/2 recipients with disease into syngeneic donor-type or allogeneic host-type tertiary recipients propagated autoimmune-like manifestations in both. Furthermore, TCR spectratyping revealed that the clonal expansion of the autoreactive CD4(+) T cells in cGVHD recipients was initiated by an alloimmune response. Finally, hybridoma CD4(+) T clones derived from DBA/2 recipients with disease proliferated similarly in response to stimulation by syngeneic donor-type or allogeneic host-type dendritic cells. These results demonstrate that the autoimmune-like manifestations in cGVHD can be mediated by a population of donor CD4(+) T cells in transplants that simultaneously recognize Ags presented by both donor and host APCs.


Subject(s)
Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Graft vs Host Disease/immunology , H-2 Antigens/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Autoantibodies/biosynthesis , Autoantigens/administration & dosage , Autoantigens/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Chronic Disease , Clone Cells , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Histocompatibility Testing , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Models, Animal , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/transplantation
14.
J Environ Qual ; 32(6): 2364-72, 2003.
Article in English | MEDLINE | ID: mdl-14674561

ABSTRACT

The high-level radioactive, Al-rich, concentrated alkaline and saline waste fluids stored in underground tanks have accidentally leaked into the vadose zone at the Hanford Site in Washington State. In addition to dissolution, precipitation is likely to occur when these waste fluids contact the sediments. The objective of this study was to investigate the solid phase transformations caused by dissolution and precipitation in the sediments treated with solutions similar to the waste fluids. Batch experiments at 323 K were conducted in metal- and glass-free systems under CO2 and O2 free conditions. Results from X-ray diffraction (XRD), quantitative X-ray diffraction (QXRD), scanning electron microscopy (SEM), energy dispersive spectroscopy (EDS), and energy dispersive X-ray fluorescence spectroscopy (EDXRF) indicated that significant solid phase transformations occurred in the sediments contacted with Al-rich, hyperalkaline, and saline solutions. The XRD and QXRD analyses confirmed that smectite and most likely biotite underwent dissolution. The SEM and the qualitative EDS analyses confirmed the formation of alumino-silicates in the groups of cancrinite and probably sodalite. The morphology of the alumino-silicates secondary phases changed in response to changes in the Si/Al aqueous molar ratio. The transformations in the sediments triggered by dissolution (weathering of soil minerals) and precipitation (formation of secondary phases with high specific surface area and probably high sorption capacities) may play a significant role in the immobilization and ultimate fate of radionuclides and contaminants such as Cs, Sr, and U in the Hanford vadose zone.


Subject(s)
Aluminum/analysis , Radioactive Waste/analysis , Soil Pollutants/analysis , Accidents , Chemical Precipitation , Humans , Hydrogen-Ion Concentration , Metals, Heavy/analysis , Sodium Chloride/analysis , Solubility , Washington , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...