Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35536669

ABSTRACT

BACKGROUNDCOVID-19 remains a global health emergency with limited treatment options, lagging vaccine rates, and inadequate healthcare resources in the face of an ongoing calamity. The disease is characterized by immune dysregulation and cytokine storm. Cyclosporine A (CSA) is a calcineurin inhibitor that modulates cytokine production and may have direct antiviral properties against coronaviruses.METHODSTo test whether a short course of CSA was safe in patients with COVID-19, we treated 10 hospitalized, oxygen-requiring, noncritically ill patients with CSA (starting at a dose of 9 mg/kg/d). We evaluated patients for clinical response and adverse events, measured serum cytokines and chemokines associated with COVID-19 hyperinflammation, and conducted gene-expression analyses.RESULTSFive participants experienced adverse events, none of which were serious; transaminitis was most common. No participant required intensive care unit-level care, and all patients were discharged alive. CSA treatment was associated with significant reductions in serum cytokines and chemokines important in COVID-19 hyperinflammation, including CXCL10. Following CSA administration, we also observed a significant reduction in type I IFN gene expression signatures and other transcriptional profiles associated with exacerbated hyperinflammation in the peripheral blood cells of these patients.CONCLUSIONShort courses of CSA appear safe and feasible in patients with COVID-19 who require oxygen and may be a useful adjunct in resource-limited health care settings.TRIAL REGISTRATIONThis trial was registered on ClinicalTrials.gov (Investigational New Drug Application no. 149997; ClinicalTrials.gov NCT04412785).FUNDINGThis study was internally funded by the Center for Cellular Immunotherapies.


Subject(s)
COVID-19 Drug Treatment , Cyclosporine/therapeutic use , Cytokines , Humans , Oxygen , SARS-CoV-2
2.
Cureus ; 13(10): e19102, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34868754

ABSTRACT

We present a systematic review and pooled analysis of clinical studies to date that (1) specifically compare the protection of natural immunity in the COVID-recovered versus the efficacy of complete vaccination in the COVID-naive, and (2) the added benefit of vaccination in the COVID-recovered, for prevention of subsequent SARS-CoV-2 infection. Using the PRISMA 2020 guidance, we first conducted a systematic review of available literature on PubMed, MedRxIV and FDA briefings to identify clinical studies either comparing COVID vaccination to natural immunity or delineating the benefit of vaccination in recovered individuals. After assessing eligibility, studies were qualitatively appraised and formally graded using the NOS system for observational, case-control and RCTs. Incidence rates were tabulated for the following groups: never infected (NI) and unvaccinated (UV), NI and vaccinated (V), previously infected (PI) and UV, PI and V. Pooling were performed by grouping the RCTs and observational studies separately, and then all studies in total. Risk ratios and differences are reported for individual studies and pooled groups, in 1) NPI/V vs PI/UV and 2) PI/UV vs PI/V analysis. In addition, the number needed to treat (NNT) analysis was performed for vaccination in naïve and previously infected cohorts. Nine clinical studies were identified, including three randomized controlled studies, four retrospective observational cohorts, one prospective observational cohort, and a case-control study. The NOS quality appraisals of these articles ranged from four to nine (out of nine stars). All of the included studies found at least statistical equivalence between the protection of full vaccination and natural immunity; and, three studies found superiority of natural immunity. Four observational studies found a statistically significant incremental benefit to vaccination in the COVID-recovered individuals. In a total pooled analysis, the incidence in NPI/V trended higher than PI/UV groups (RR=1.86 [95%CI 0.77-4.51], P=0.17). Vaccination in COVID-recovered individuals provided modest protection from reinfection (RR=1.82 [95%CI 1.21-2.73], P=0.004), but the absolute risk difference was extremely small (AR= 0.004 person-years [95% CI 0.001-0.007], P=0.02). The NNT to prevent one annual case of infection in COVID-recovered patients was 218, compared to 6.5 in COVID-naïve patients, representing a 33.5-fold difference in benefit between the two populations. COVID-recovered individuals represent a distinctly different benefit-risk calculus. While vaccinations are highly effective at protecting against infection and severe COVID-19 disease, our review demonstrates that natural immunity in COVID-recovered individuals is, at least, equivalent to the protection afforded by complete vaccination of COVID-naïve populations. There is a modest and incremental relative benefit to vaccination in COVID-recovered individuals; however, the net benefit is marginal on an absolute basis. Therefore, vaccination of COVID-recovered individuals should be subject to clinical equipoise and individual preference.

5.
J Surg Res ; 187(1): 310-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24267616

ABSTRACT

BACKGROUND: Long-lived plasma cells (PCs) that form after alloantigen sensitization produce donor-specific alloantibodies that generate a positive serum crossmatch and preclude transplantation. New approaches for desensitization, including PC depletion with proteasome inhibition, show promise but carry considerable toxicity. Recently, eosinophils have been shown to govern PC persistence. Interleukin 5 (IL-5) depletion is known to reduce eosinophils in human asthmatics. We hypothesized that treatment with an anti-IL-5 antibody can deplete alloreactive PCs, reduce donor-specific alloantibodies, and serve as a less toxic alternative to proteasome inhibition. METHODS: BALB/c mice were sensitized with B6 skin allografts. Starting at 8 wk after sensitization, control mice received injections of phosphate-buffered saline, whereas experimental mice received weekly injections of an anti-IL-5 antibody. PCs were enumerated by enzyme-linked immunosorbent spot after 8 wk. RESULTS: All control and experimental recipients of skin allografts developed positive crossmatches when screened at 8 wk after sensitization. All experimental mice treated with anti-IL-5 showed a reduction in their total PC numbers. Also, in contrast to the known adverse effects of proteasome inhibition, experimental mice treated with anti-IL-5 exhibited negligible weight loss or lymphopenia. CONCLUSIONS: Treatment with anti-IL-5 is sufficient to reduce, but not eliminate, alloreactive PCs in the bone marrow. This is because of the targeted reduction of eosinophils leading to a reduction in the PC survival factors a proliferation-inducing ligand and IL-6. Generalized toxicity was not observed in experimental mice. Overall, IL-5 directed immunotherapy can eliminate PC's but is unlikely to be a clinically significant desensitization strategy given the persistence of DSA.


Subject(s)
Antibodies/pharmacology , Desensitization, Immunologic/methods , Immunotherapy/methods , Interleukin-5/antagonists & inhibitors , Plasma Cells/immunology , Skin Transplantation , Animals , Antibodies/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , Eosinophils/cytology , Eosinophils/immunology , Interleukin-5/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Isoantibodies/immunology , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasma Cells/cytology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
7.
Transplantation ; 93(7): 676-85, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22262127

ABSTRACT

BACKGROUND: Immunologic rejection is a major barrier to successful long-term outcomes in clinical transplantation. The importance of B lymphocytes-and their secretory products, alloantibodies-in the pathogenesis of allograft rejection is accepted. Furthermore, it is now clear that the dominant regulator of peripheral B-cell homeostasis and tolerance is the B-lymphocyte stimulator (BLyS), also referred to as the B-cell activating factor (BAFF). Recently, a novel class of clinical immunotherapeutic agents specific for BLyS, and its family of cytokines, has emerged for the treatment of B-cell-mediated diseases. In this study, we demonstrate the potential utility of BLyS-directed immunotherapy in preventing allograft rejection using a murine islet transplantation model. METHODS: A transient period of mature peripheral B-cell depletion was induced by means of in vivo BLyS neutralization using a murine analog of the monoclonal antibody, Benlysta. Subsequently, fully major histocompatibility complex-mismatched islets were transplanted into naïve diabetic mice followed by a short course of rapamycin. RESULTS: After BLyS neutralization, indefinite islet allograft survival was achieved. Induction therapy with rapamycin was necessary, but not sufficient, for the achievement of this long-term graft survival. The tolerant state was associated with (1) abrogation of the donor-specific antibody response, (2) transient preponderance of immature/transitional B cells in all lymphoid organs, (3) impaired CD4 T-cell activation during the period of B-cell depletion, and (4) presence of a "regulatory" cytokine milieu. CONCLUSIONS: In vivo BLyS neutralization effectively induces humoral tolerance and promotes long-term islet allograft survival in mice. Therefore, B-lymphocyte-directed immunotherapy targeting the homeostatic regulator, BLyS, may be effective in promoting transplantation tolerance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , B-Cell Activating Factor/antagonists & inhibitors , B-Lymphocytes/drug effects , Diabetes Mellitus, Experimental/surgery , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Transplantation Tolerance/drug effects , Animals , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Graft Rejection/immunology , Histocompatibility/drug effects , Immunity, Humoral/drug effects , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Sirolimus/pharmacology , Time Factors , Transplantation, Homologous
8.
Semin Immunol ; 24(2): 109-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21978627

ABSTRACT

The current mainstay of immunotherapy in clinical transplantation is T lymphocyte directed. However, it has long been appreciated that the emergence of an alloimmune response mounted by the B lymphocyte compartment and detectable as donor-specific antibodies is a critical challenge to long-term graft survival. Thus, achieving robust transplantation tolerance will require induction of tolerance in both the T- and B-cell compartments. Here we propose that the natural developmental propensity of the B-lymphocyte compartment acquisition of tolerance to self-antigens can be recapitulated to achieve humoral transplantation tolerance. It is our contention B-lymphocyte directed induction immunotherapy would be an important component of emerging strategies for induction of Transplantation tolerance.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocyte Subsets/cytology , Isoantibodies/biosynthesis , Isoantigens/immunology , Transplantation Tolerance/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/transplantation , Humans , Immunotherapy/methods , Isoantibodies/immunology , Lymphocyte Depletion/methods
9.
Clin Transplant ; 26(3): 489-94, 2012.
Article in English | MEDLINE | ID: mdl-22032287

ABSTRACT

BACKGROUND: ABO compatibility creates a disadvantage for O and B renal allograft candidates. A2 ABO incompatible transplant may decrease waiting times and generate equivalent graft survival to an ABO compatible transplant. METHODS: Death-censored graft survival was compared between A recipients and O, B, and AB recipients of an A2 allograft with multivariate Cox regression models utilizing data from the United Network of Organ Sharing (UNOS) between 1997 and 2007. RESULTS: Eighty-five percent of A2 kidneys were transplanted into ABO compatible recipients vs. 15% into ABO incompatible recipients. Rates of A2 incompatible kidney transplants did not increase over the study period (14.8% to 14.6%). Mean wait time for A2→O kidneys was 337 vs. 684 d for O→O and for A2→B kidneys, 542 vs. 734 d for B→B. Adjusted relative risk of graft loss at five-yr was similar between O, B, and AB recipients compared to A recipients of an A2 allograft, corresponding to a five-yr graft survival of 84%, 86.2%, 86.1%, and 86.1%, respectively. CONCLUSION: A2 incompatible kidney transplantation is underutilized. Graft outcomes are similar among A2 compatible and incompatible recipients. Shorter waiting time and improved access might be achieved if A2 kidneys are considered in all blood groups.


Subject(s)
ABO Blood-Group System/immunology , Blood Group Incompatibility/immunology , Graft Survival/immunology , Kidney Transplantation/immunology , Kidney Transplantation/statistics & numerical data , Transplantation Immunology , Adult , Female , Follow-Up Studies , Humans , Kidney Failure, Chronic , Kidney Transplantation/mortality , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Tissue Donors , Tissue and Organ Procurement
10.
Curr Opin Immunol ; 23(5): 685-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21982511

ABSTRACT

T lymphocytes are the primary targets of immunotherapy in clinical transplantation. However, B lymphocytes are detrimental to graft survival by virtue of their capacity to present antigen to T cells via the indirect pathway of allorecognition and the generation of donor specific alloantibody. Furthermore, the long-term survival of organ allografts remains challenged by chronic rejection, a process in which activated B cells have been found to play a significant role. Therefore, the achievement of transplantation tolerance will likely require induction of both T and B cell tolerance to alloantigens. Moreover, human and animal investigations have shown that subsets of B cells, Transitional and Regulatory, are inherently tolerogenic. Developing therapeutic strategies that exploit these populations may be key to achieving transplantation tolerance. In this review we describe the current evidence for the essential role of B cells in transplant tolerance and discuss emerging B cell directed strategies to achieve allograft tolerance.


Subject(s)
Adaptive Immunity , B-Lymphocytes/immunology , Graft Rejection/prevention & control , Heart Transplantation , Immunotherapy/methods , Isoantigens/immunology , Transplantation Tolerance , Animals , Antigen Presentation , B-Lymphocytes/classification , B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Graft Rejection/immunology , Graft Survival/immunology , Humans , Isoantibodies/immunology , Mice , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology
11.
Immunol Res ; 51(1): 1-4, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21948256

ABSTRACT

Transplantation tolerance remains an elusive goal as B-cell-initiated chronic humoral rejection evades current immunosuppression. B-cell-directed therapy is thus emerging as a key component in achieving transplantation tolerance and long-term graft survival. Here, we propose strategies of B-cell repertoire remodeling to achieve humoral unresponsiveness to donor antigens with implementation of fundamental B-cell immunobiology and use of newly developed B-cell-directed agents.


Subject(s)
B-Lymphocytes/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Models, Immunological , Transplantation Tolerance/immunology , Animals , B-Lymphocytes/pathology , Graft Rejection/pathology , Humans
12.
J Immunol ; 186(1): 614-20, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21084661

ABSTRACT

A major obstacle to transplantation tolerance is humoral immunity. In this paper, we demonstrate that the intrinsic developmental propensity of the B lymphocyte compartment for acquisition of self-tolerance can be harnessed to induce humoral unresponsiveness to transplanted alloantigens. In the current study, when transitional B cells developed in the presence of donor lymphoid cells, the mature B lymphocyte compartment failed to mount a donor-specific alloantibody response to an organ transplant--despite unrestrained acute T cell-mediated allograft rejection. Specifically, we generated an experimental system wherein a B6 strain B cell compartment developed de novo in the presence of F1 (B6xBALB/c) lymphoid cells and in a T cell-deficient setting. Following establishment of a steady-state B cell compartment, these B6 mice were transplanted with heterotopic cardiac allografts from allogeneic BALB/c donors. The mice were then inoculated with purified syngeneic B6 T cells. As expected, all cardiac allografts were acutely rejected. However, the B lymphocyte compartment of these mice was completely inert in its capacity to form a BALB/c-specific alloantibody response. Using an alloantigen-specific Ig transgenic system, we demonstrated that this profound degree of humoral tolerance was caused by clonal deletion of alloreactive specificities from the primary B cell repertoire. Thus, de novo B cell compartment development at the time of transplantation is of critical importance in recipient repertoire "remodeling" to a humoral tolerant state.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Isoantibodies/biosynthesis , Transplantation Tolerance , Adoptive Transfer , Animals , Antibody Specificity/genetics , B-Lymphocyte Subsets/transplantation , Bone Marrow Transplantation/immunology , Cell Differentiation/genetics , Clone Cells , Heart Transplantation/immunology , Hematopoietic Stem Cell Transplantation , Isoantigens/genetics , Isoantigens/immunology , Lymphocyte Depletion , Lymphocyte Transfusion , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Skin Transplantation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Transplantation Tolerance/genetics
13.
Transplant Rev (Orlando) ; 24(4): 207-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20655723

ABSTRACT

Current strategies for immunotherapy after transplantation are primarily T-lymphocyte directed and effectively abrogate acute rejection. However, the reality of chronic allograft rejection attests to the fact that transplantation tolerance remains an elusive goal. Donor-specific antibodies are considered the primary cause of chronic rejection. When naive, alloreactive B-cells encounter alloantigen and are activated, a resilient "sensitized" state, characterized by the presence of high-affinity antibody, is established. Here, we will delineate findings that support transient B-lymphocyte depletion therapy at the time of transplantation to preempt sensitization by eliminating alloreactive specificities from the recipient B-cell pool (ie, "repertoire remodeling"). Recent advances in our understanding of B-lymphocyte homeostasis provide novel targets for immunomodulation in transplantation. Specifically, the tumor necrosis factor-related cytokine BLyS is the dominant survival factor for "tolerance-susceptible" transitional and "preimmune" mature follicular B-cells. The transitional phenotype is the intermediate through which all newly formed B-cells pass before maturing into the follicular subset, which is responsible for mounting an alloantigen-specific antibody response. Systemic BLyS levels dictate the stringency of negative selection during peripheral B-cell repertoire development. Thus, targeting BLyS will likely provide an opportunity for repertoire-directed therapy to eliminate alloreactive B-cell specificities in transplant recipients, a requirement for the achievement of humoral tolerance and prevention of chronic rejection. In this review, the fundamentals of preimmune B-cell selection, homeostasis, and activation will be described. Furthermore, new and current B-lymphocyte-directed therapy for antibody-mediated rejection and the highly sensitized state will be discussed. Overall, our objective is to propose a rational approach for induction of humoral transplantation tolerance by remodeling the primary B-cell repertoire of the allograft recipient.


Subject(s)
B-Cell Activating Factor/therapeutic use , B-Lymphocytes/immunology , Immunotherapy/methods , Transplantation Immunology/physiology , Autoantigens/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Homeostasis , Humans , Immune Tolerance/immunology , Isoantibodies/immunology , Isoantigens/immunology , T-Lymphocytes/immunology
14.
Immunogenetics ; 62(6): 397-407, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20387063

ABSTRACT

In the present study, we characterize a polymorphism in the CD93 molecule, originally identified as the receptor for the C1q complement component (i.e., C1qRp, or AA4.1) in non-obese diabetic (NOD) mice. This allele carries a coding polymorphism in the first epidermal growth factor-like domain of CD93, which results in an amino acid substitution from Asn-->His at position 264. This polymorphism does not appear to influence protein translation or ecto-domain cleavage, as CD93 is detectable in bone-marrow-derived macrophage and B-cell precursor lysates and in soluble form in the serum. The NOD CD93 isoform causes a phenotypic aberrancy in the early B-cell developmental stages (i.e., pro-, pre-, immature, and transitional), likely related to a conformational variation. Interestingly, the NZB/W F1 strain, which serves as a murine model of Lupus, also expresses an identical CD93 sequence polymorphism. Cd93 is located within the NOD Idd13 locus and is also tightly linked to the NZB/W F1 Wbw1 and Nkt2 disease susceptibility loci, which are thought to regulate natural killer T (NKT) cell homeostasis. Consistent with this genetic linkage, we found B6 CD93(-/-) and B6.NOD(Idd13) mice to be susceptible to a profound CD4(+) NKT cell deficient state. These data suggest that Cd93 may be an autoimmune susceptibility gene residing within the Idd13 locus, which plays a role in regulating absolute numbers of CD4(+) NKT cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Membrane Glycoproteins/genetics , Natural Killer T-Cells/immunology , Polymorphism, Genetic , Receptors, Complement/genetics , Animals , Base Sequence , Chromosome Mapping , Lymphocyte Count , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred NZB , Molecular Sequence Data , Receptors, Complement/physiology
15.
Expert Rev Clin Immunol ; 5(6): 703, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20161663

ABSTRACT

T lymphocytes are the primary targets of immunotherapy in clinical transplantation; however, B lymphocytes and their secreted alloantibodies are also highly detrimental to the allograft. Therefore, the achievement of sustained organ transplant survival will likely require the induction of B-lymphocyte tolerance. During development, acquisition of B-cell tolerance to self-antigens relies on clonal deletion in the early stages of B-cell compartment ontogeny. We contend that this mechanism should be recapitulated in the setting of alloantigens and organ transplantation to eliminate the alloreactive B-cell subset from the recipient. Clinically feasible targets of B-cell-directed immunotherapy, such as CD20 and B-lymphocyte stimulator (BLyS), should drive upcoming clinical trials aimed at remodeling the recipient B-cell repertoire.

16.
J Immunol ; 181(11): 8133-44, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19018006

ABSTRACT

B lymphocytes are required for the pathogenesis of autoimmune diabetes in NOD mice. Previous studies established that a lymphopenic transitional (TR) B cell compartment reduces the competitive constraint on the entry of newly emerging TR B cells into the splenic follicle (FO), thereby disrupting a peripheral negative selection checkpoint in NOD mice. Thus, development of clinically feasible immunotherapeutic approaches for restoration of appropriate negative selection is essential for the prevention of anti-islet autoimmunity. In this study we hypothesized that in vivo neutralization of the B lymphocyte stimulator (BLyS/BAFF) may enhance the stringency of TR-->FO selection by increasing TR B cell competition for follicular entry in NOD mice. This study demonstrated that in vivo BLyS neutralization therapy leads to the depletion of follicular and marginal zone B lymphocytes. Long-term in vivo BLyS neutralization caused an increased TR:FO B cell ratio in the periphery indicating a relative resistance to follicular entry. Moreover, in vivo BLyS neutralization: 1) restored negative selection at the TR-->FO checkpoint, 2) abrogated serum insulin autoantibodies, 3) reduced the severity of islet inflammation, 4) significantly reduced the incidence of spontaneous diabetes, 5) arrested the terminal stages of islet cell destruction, and 6) disrupted CD4 T cell activation in NOD mice. Overall, this study demonstrates the efficacy of B lymphocyte-directed therapy via in vivo BLyS neutralization for the prevention of autoimmune diabetes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Autoimmunity/drug effects , B-Cell Activating Factor/antagonists & inhibitors , Diabetes Mellitus, Type 1/drug therapy , Immunotherapy , Insulin-Secreting Cells/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Autoantibodies/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Insulin/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD
17.
Nat Med ; 13(11): 1295-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17965721

ABSTRACT

We found that an induction immunotherapy regimen consisting of rabbit anti-thymocyte globulin (Thymoglobulin) and the monoclonal antibody to CD20 rituximab (Rituxan) promoted long-term islet allograft survival in cynomolgus macaques maintained on rapamycin monotherapy. B lymphocyte reconstitution after rituximab-mediated depletion was characterized by a preponderance of immature and transitional cells, whose persistence was associated with long-term islet allograft survival. Development of donor-specific alloantibodies was abrogated only in the setting of continued rapamycin monotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B-Lymphocyte Subsets/immunology , Graft Survival/immunology , Immunotherapy, Active , Islets of Langerhans Transplantation/immunology , Animals , Antibodies, Monoclonal, Murine-Derived , Antilymphocyte Serum , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Immunotherapy, Active/methods , Lymphocyte Depletion , Macaca fascicularis , Rituximab , Transplantation, Homologous
18.
J Immunol ; 177(11): 7715-22, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17114442

ABSTRACT

Acute allograft rejection requires the activation of alloreactive CD4 T cells. Despite the capacity of B cells to act as potent APCs capable of activating CD4 T cells in vivo, their role in the progression of acute allograft rejection was unclear. To determine the contribution of B cell APC function in alloimmunity, we engineered mice with a targeted deficiency of MHC class II-mediated Ag presentation confined to the B cell compartment. Cardiac allograft survival was markedly prolonged in these mice as compared to control counterparts (median survival time, >70 vs 9.5 days). Mechanistically, deficient B cell-mediated Ag presentation disrupted both alloantibody production and the progression of CD4 T cell activation following heart transplantation. These findings demonstrate that indirect alloantigen presentation by recipients' B cells plays an important role in the efficient progression of acute vascularized allograft rejection.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Lymphocyte Activation/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Isoantibodies/blood , Isoantibodies/immunology , Isoantigens/immunology , Mice , Transplantation Chimera
19.
J Immunol ; 176(12): 7159-64, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16751358

ABSTRACT

Developing B cells undergo selection at multiple checkpoints to eliminate autoreactive clones. We analyzed B cell kinetics in the NOD mouse to establish whether these checkpoints are intact. Our results show that although bone marrow production is normal in NOD mice, transitional (TR) B cell production collapses at 3 wk of age, reflecting a lack of successful immature B cell migration to the periphery. This yields delayed establishment of the follicular pool and a lack of selection at the TR checkpoint, such that virtually all immature B cells that exit the bone marrow mature without further selection. These findings suggest that compromised TR B cell generation in NOD mice yields relaxed TR selection, affording autoreactive specificities access to mature pools.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/pathology , Cell Cycle/immunology , Cell Differentiation/immunology , Aging/genetics , Aging/immunology , Animals , B-Lymphocyte Subsets/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cell Migration Inhibition , Clone Cells , Genetic Predisposition to Disease , Lymphocyte Count , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Species Specificity , Stem Cells/cytology , Stem Cells/immunology
20.
Immunology ; 116(3): 400-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16236130

ABSTRACT

The present study aimed to determine whether the frequency of double positive (DP) thymocytes expressing alphabeta T-cell receptor (TCR) clonotypes at the time of selection regulates peripheral CD4 T-cell compartment size. Scid recipients were inoculated with various ratios of TCR Calpha(0/0) and wild-type bone marrow (BM) stem cells. Increasing the frequency of TCR Calpha(0/0) thymocytes at steady-state introduced a graded decrease in the maturation probability of the total DP thymocyte pool. At 12-14 weeks following BM inoculation, the frequency of TCR Calpha(0/0) DP thymocytes was inversely correlated with that of CD4 single positive (SP) thymocytes. Notwithstanding, a decreased frequency of wild-type DP thymocytes led to a marked increase in their transit efficiency from the DP to SP compartments. The frequency-dependent increase in thymocyte transit efficiency was associated with a CD4 SP cell surface phenotype indicative of increased antigenic experience. Importantly, the frequency of DP thymocytes capable of expressing TCR clonotypes dictated the steady-state size of the peripheral CD4 T cell compartment and its potential for homeostatic proliferation. Collectively, these results indicate that the efficiency of DP to CD4 SP transit is a frequency dependent process, which determines (1) the steady-state size of the peripheral T cell compartment and (2) the threshold for homeostatic expansion of peripheral CD4 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Bone Marrow Transplantation , Cell Differentiation/immunology , Cell Proliferation , Flow Cytometry , Homeostasis/immunology , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, SCID , Transplantation Chimera/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...