Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Trends Immunol ; 45(1): 48-61, 2024 01.
Article in English | MEDLINE | ID: mdl-38123369

ABSTRACT

In the quest for more precise and effective organ transplantation therapies, chimeric antigen receptor (CAR) regulatory T cell (Treg) therapies represent a potential cutting-edge advance. This review comprehensively analyses CAR Tregs and how they may address important drawbacks of polyclonal Tregs and conventional immunosuppressants. We examine a growing body of preclinical findings of CAR Treg therapy in transplantation, discuss CAR Treg design specifics, and explore established and attractive new targets in transplantation. In addition, we explore present impediments where future studies will be necessary to determine the efficacy of CAR Tregs in reshaping alloimmune responses and transplant microenvironments to reduce reliance on chemical immunosuppressants. Overall, ongoing studies and trials are crucial for understanding the full scope of CAR Treg therapy in transplantation.


Subject(s)
Organ Transplantation , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive , Immunosuppressive Agents , T-Lymphocytes, Regulatory , Receptors, Antigen, T-Cell
2.
J Infect Dis ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38513368

ABSTRACT

BACKGROUND: Immunosuppressed individuals, including solid organ transplant recipients (SOTRs), are at high risk for severe COVID-19. METHODS: This open-label, phase 3b trial evaluated mRNA-1273 in 137 adult kidney and 77 liver SOTRs and 20 immunocompetent participants. In Part A, SOTRs received three 100-µg doses of mRNA-1273; immunocompetent participants received 2 doses. In Part B, an additional 100-µg dose was offered ≥4 months post-primary series. Here, we report interim trial results. RESULTS: mRNA-1273 was well-tolerated in SOTRs. Four serious adverse events were considered vaccine-related by the investigator in 3 SOTRs with pre-existing comorbidities. No vaccine-related biopsy-proven organ rejection events or deaths were reported. mRNA-1273 elicited modest neutralizing antibody (nAb) responses after dose 2 and improved responses after dose 3 in SOTRs. Post-dose 3 responses among liver SOTRs were comparable to post-dose 2 responses in immunocompetent participants. Post-additional dose responses were increased in SOTRs regardless of the primary series vaccination. In liver SOTRs, post-additional dose responses were ∼3-fold higher versus post-dose 2 but were lower than immunocompetent participant responses. Most kidney SOTRs received multiple immunosuppressants and had reduced antibody responses versus liver SOTRs. CONCLUSIONS: mRNA-1273 (100 µg) was well-tolerated and dose 3 and the additional dose improved antibody responses among SOTRs.

3.
J Am Soc Nephrol ; 34(5): 846-856, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36758118

ABSTRACT

SIGNIFICANCE STATEMENT: Donor-specific antibodies against class II HLA are a major cause of chronic kidney graft rejection. Nonetheless, some patients presenting with these antibodies remain in stable histological and clinical condition. This study describes the use of endothelial colony-forming cell lines to test the hypothesis of the heterogeneous expression of HLA molecules on endothelial cells in humans. Flow cytometry and immunofluorescence staining revealed substantial interindividual and interlocus variability, with HLA-DQ the most variable. Our data suggest that the expression of HLA class II is predicted by locus. The measurement of endothelial expression of HLA class II in the graft could present a novel paradigm in the evaluation of the alloimmune risk in transplantation and certain diseases. BACKGROUND: HLA antigens are important targets of alloantibodies and allospecific T cells involved in graft rejection. Compared with research into understanding alloantibody development, little is known about the variability in expression of their ligands on endothelial cells. We hypothesized individual variability in the expression of HLA molecules. METHODS: We generated endothelial colony forming cell lines from human peripheral blood mononuclear cells ( n =39). Flow cytometry and immunofluorescence staining were used to analyze the cells, and we assessed the relationship between HLA-DQ expression and genotype. Two cohorts of kidney transplant recipients were analyzed to correlate HLA-DQ mismatches with the extent of intragraft microvascular injury. RESULTS: Large variability was observed in the expression of HLA class II antigens, not only between individuals but also between subclasses. In particular, HLA-DQ antigens had a low and heterogeneous expression, ranging from 0% to 85% positive cells. On a within-patient basis, this expression was consistent between endothelial cell colonies and antigen-presenting cells. HLA-DQ5 and -DQ6 were associated with higher levels of expression, whereas HLA-DQ7, -DQ8, and -DQ9 with lower. HLA-DQ5 mismatches among kidney transplant recipients were associated with significant increase in graft microvascular. CONCLUSION: These data challenge the current paradigm that HLA antigens, in particular HLA class II, are a single genetic and post-translational entity. Understanding and assessing the variability in the expression of HLA antigens could have clinical monitoring and treatment applications in transplantation, autoimmune diseases, and oncology.


Subject(s)
Endothelial Cells , Kidney Transplantation , Humans , Leukocytes, Mononuclear , HLA Antigens , HLA-DQ Antigens , Isoantibodies , Graft Rejection , Histocompatibility Antigens Class II , Graft Survival
4.
Proc Natl Acad Sci U S A ; 117(11): 6042-6046, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32111690

ABSTRACT

Induction of longstanding immunologic tolerance is essential for survival of transplanted organs and tissues. Despite recent advances in immunosuppression protocols, allograft damage inflicted by antibody specific for donor organs continues to represent a major obstacle to graft survival. Here we report that activation of regulatory CD8 T cells (CD8 Treg) that recognize the Qa-1 class Ib major histocompatibility complex (MHC), a mouse homolog of human leukocyte antigen-E (HLA-E), inhibits antibody-mediated immune rejection of heart allografts. We analyzed this response using a mouse model that harbors a point mutation in the class Ib MHC molecule Qa-1, which disrupts Qa-1 binding to the T cell receptor (TCR)-CD8 complex and impairs the CD8 Treg response. Despite administration of cytotoxic T lymphocyte antigen 4 (CTLA-4) immunoglobulin (Ig), Qa-1 mutant mice developed robust donor-specific antibody responses and accelerated heart graft rejection. We show that these allo-antibody responses reflect diminished Qa-1-restricted CD8 Treg-mediated suppression of host follicular helper T cell-dependent antibody production. These findings underscore the critical contribution of this Qa-1/HLA-E-dependent regulatory pathway to maintenance of transplanted organs and suggest therapeutic approaches to ameliorate allograft rejection.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/adverse effects , Histocompatibility Antigens Class I/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , Allografts/immunology , Allografts/metabolism , Animals , Disease Models, Animal , Graft Rejection/blood , Graft Rejection/genetics , Graft Survival/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Tolerance , Isoantibodies/immunology , Isoantibodies/metabolism , Isoantigens/immunology , Isoantigens/metabolism , Mice , Myocardium/immunology , Myocardium/metabolism , Point Mutation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Transplantation, Homologous/adverse effects
5.
Am J Transplant ; 22(12): 3130-3136, 2022 12.
Article in English | MEDLINE | ID: mdl-35727916

ABSTRACT

The neutralizing monoclonal antibody combination of tixagevimab/cilgavimab has been shown to reduce the risk of SARS-CoV-2 infection in unvaccinated individuals during the Alpha (B.1.1.7) and Delta (B.1.617.2) waves. However, data on the efficacy and safety of tixagevimab/cilgavimab in vaccinated solid organ transplant recipients during the Omicron wave is limited. To address this, we conducted a retrospective cohort study comparing 222 solid organ transplant recipients (SOTRs) who received tixagevimab/cilgavimab for pre-exposure prophylaxis and 222 vaccine-matched solid organ transplant recipients who did not receive tixagevimab/cilgavimab. Breakthrough SARS-CoV-2 infections occurred in 11 (5%) of SOTRs who received tixagevimab/cilgavimab and in 32 (14%) of SOTRs in the control group (p < .001). In the tixagevimab/cilgavimab group, SOTRs who received the 150-150 mg dose had a higher incidence of breakthrough infections compared to those who received the 300-300 mg dose (p = .025). Adverse events were uncommon, occurring in 4% of our cohort and most were mild. There was no significant change in serum creatinine or liver chemistries in kidney and liver transplant recipients, respectively. In conclusion, we found that tixagevimab/cilgavimab use is safe and associated with a lower risk of breakthrough SARS-CoV-2 infection in vaccinated solid organ transplant recipients during the Omicron wave.


Subject(s)
COVID-19 , Organ Transplantation , Pre-Exposure Prophylaxis , Humans , SARS-CoV-2 , Retrospective Studies , Antibodies, Monoclonal , Transplant Recipients , Organ Transplantation/adverse effects
6.
Am J Transplant ; 22 Suppl 4: 45-57, 2022 12.
Article in English | MEDLINE | ID: mdl-36453708

ABSTRACT

Of all kidney transplants, half are still lost in the first decade after transplantation. Here, using genetics, we probed whether interleukin 6 (IL-6) could be a target in kidney transplantation to improve graft survival. Additionally, we investigated if a genetic risk score (GRS) based on IL6 and IL10 variants could improve prognostication of graft loss. In a prospective cohort study, DNA of 1271 donor-recipient kidney transplant pairs was analyzed for the presence of IL6, IL6R, IL10, IL10RA, and IL10RB variants. These polymorphisms and their GRS were then associated with 15-year death-censored allograft survival. The C|C-genotype of the IL6 polymorphism in donor kidneys and the combined C|C-genotype in donor-recipient pairs were both associated with a reduced risk of graft loss (p = .043 and p = .042, respectively). Additionally, the GRS based on IL6, IL6R, IL10, IL10RA, and IL10RB variants was independently associated with the risk of graft loss (HR 1.53, 95%-CI [1.32-1.84]; p < .001). Notably, the GRS improved risk stratification and prediction of graft loss beyond the level of contemporary clinical markers. Our findings reveal the merits of a polygenic IL-6-based risk score strengthened with IL-10- polymorphisms for the prognostication and risk stratification of late graft failure in kidney transplantation.


Subject(s)
Interleukin-10 , Interleukin-6 , Humans , Interleukin-10/genetics , Interleukin-6/genetics , Prospective Studies , Kidney , Risk Factors , Allografts
7.
J Am Soc Nephrol ; 32(4): 994-1004, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33658284

ABSTRACT

BACKGROUND: Developing a noninvasive clinical test to accurately diagnose kidney allograft rejection is critical to improve allograft outcomes. Urinary exosomes, tiny vesicles released into the urine that carry parent cells' proteins and nucleic acids, reflect the biologic function of the parent cells within the kidney, including immune cells. Their stability in urine makes them a potentially powerful tool for liquid biopsy and a noninvasive diagnostic biomarker for kidney-transplant rejection. METHODS: Using 192 of 220 urine samples with matched biopsy samples from 175 patients who underwent a clinically indicated kidney-transplant biopsy, we isolated urinary exosomal mRNAs and developed rejection signatures on the basis of differential gene expression. We used crossvalidation to assess the performance of the signatures on multiple data subsets. RESULTS: An exosomal mRNA signature discriminated between biopsy samples from patients with all-cause rejection and those with no rejection, yielding an area under the curve (AUC) of 0.93 (95% CI, 0.87 to 0.98), which is significantly better than the current standard of care (increase in eGFR AUC of 0.57; 95% CI, 0.49 to 0.65). The exosome-based signature's negative predictive value was 93.3% and its positive predictive value was 86.2%. Using the same approach, we identified an additional gene signature that discriminated patients with T cell-mediated rejection from those with antibody-mediated rejection (with an AUC of 0.87; 95% CI, 0.76 to 0.97). This signature's negative predictive value was 90.6% and its positive predictive value was 77.8%. CONCLUSIONS: Our findings show that mRNA signatures derived from urinary exosomes represent a powerful and noninvasive tool to screen for kidney allograft rejection. This finding has the potential to assist clinicians in therapeutic decision making.

8.
Circulation ; 140(10): 846-863, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31266349

ABSTRACT

BACKGROUND: Transplantation is the treatment of choice for many patients with end-stage organ disease. Despite advances in immunosuppression, long-term outcomes remain suboptimal, hampered by drug toxicity and immune-mediated injury, the leading cause of late graft loss. The development of therapies that promote regulation while suppressing effector immunity is imperative to improve graft survival and minimize conventional immunosuppression. Notch signaling is a highly conserved pathway pivotal to T-cell differentiation and function, rendering it a target of interest in efforts to manipulate T cell-mediated immunity. METHODS: We investigated the pattern of Notch-1 expression in effector and regulatory T cells (Tregs) in both murine and human recipients of a solid-organ transplant. Using a selective human anti-Notch-1 antibody (aNotch-1), we examined the effect of Notch-1 receptor inhibition in full major histocompatibility complex-mismatch murine cardiac and lung transplant models, and in a humanized skin transplant model. On the basis of our findings, we further used a genetic approach to investigate the effect of selective Notch-1 inhibition in Tregs. RESULTS: We observed an increased proportion of Tregs expressing surface and intracellular (activated) Notch-1 in comparison with conventional T cells, both in mice with transplants and in the peripheral blood of patients with transplants. In the murine cardiac transplant model, peritransplant administration of aNotch-1 (days 0, 2, 4, 6, 8, and 10) significantly prolonged allograft survival in comparison with immunoglobulin G-treated controls. Similarly, aNotch-1 treatment improved both histological and functional outcomes in the murine lung transplant model. The use of aNotch-1 resulted in a reduced proportion of both splenic and intragraft conventional T cells, while increasing the proportion of Tregs. Furthermore, Tregs isolated from aNotch-1-treated mice showed enhanced suppressive function on a per-cell basis, confirmed with selective Notch-1 deletion in Tregs (Foxp3EGFPCreNotch1fl/fl). Notch-1 blockade inhibited the mammalian target of rapamycin pathway and increased the phosphorylation of STAT5 (signal transducer and activator of transcription 5) in murine Tregs. Notch-1low Tregs isolated from human peripheral blood exhibited more potent suppressive capacity than Notch-1high Tregs. Last, the combination of aNotch-1 with costimulation blockade induced long-term tolerance in a cardiac transplant model, and this tolerance was dependent on CTLA-4 (cytotoxic T-lymphocyte-associated antigen-4) signaling. CONCLUSIONS: Our data reveal a promising, clinically relevant approach for immune modulation in transplantation by selectively targeting Notch-1.


Subject(s)
Graft Rejection/metabolism , Receptor, Notch1/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Blocking/pharmacology , Cells, Cultured , Gene Expression Regulation , Graft Rejection/immunology , Graft Rejection/mortality , Humans , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Transplantation , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Survival Analysis
9.
Clin Transplant ; 34(11): e14059, 2020 11.
Article in English | MEDLINE | ID: mdl-32762055

ABSTRACT

An unprecedented global pandemic caused by a novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has quickly overwhelmed the health care systems worldwide. While there is an absence of consensus among the community in how to manage solid organ transplant recipients and donors, a platform provided by the American Society of Transplantation online community "Outstanding Questions in Transplantation," hosted a collaborative multicenter, multinational discussions to share knowledge in a rapidly evolving global situation. Here, we present a summary of the discussion in addition to the latest published literature.


Subject(s)
COVID-19 , Organ Transplantation , Pandemics , Postoperative Complications , SARS-CoV-2 , COVID-19/epidemiology , COVID-19/immunology , COVID-19/therapy , Global Health , Graft Rejection/prevention & control , Humans , Immunocompromised Host , Immunosuppressive Agents/therapeutic use , International Cooperation , Postoperative Complications/epidemiology , Postoperative Complications/immunology , Postoperative Complications/therapy , Societies, Medical
10.
Proc Natl Acad Sci U S A ; 113(52): E8425-E8432, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956634

ABSTRACT

Constitutive proteasomes (c-20S) are ubiquitously expressed cellular proteases that degrade polyubiquitinated proteins and regulate cell functions. An isoform of proteasome, the immunoproteasome (i-20S), is highly expressed in human T cells, dendritic cells (DCs), and B cells, suggesting that it could be a potential target for inflammatory diseases, including those involving autoimmunity and alloimmunity. Here, we describe DPLG3, a rationally designed, noncovalent inhibitor of the immunoproteasome chymotryptic subunit ß5i that has thousands-fold selectivity over constitutive ß5c. DPLG3 suppressed cytokine release from blood mononuclear cells and the activation of DCs and T cells, diminished accumulation of effector T cells, promoted expression of exhaustion and coinhibitory markers on T cells, and synergized with CTLA4-Ig to promote long-term acceptance of cardiac allografts across a major histocompatibility barrier. These findings demonstrate the potential value of using brief posttransplant immunoproteasome inhibition to entrain a long-term response favorable to allograft survival as part of an immunomodulatory regimen that is neither broadly immunosuppressive nor toxic.


Subject(s)
Graft Survival , Heart Transplantation/methods , Immunosuppressive Agents/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation , Cytokines/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Hep G2 Cells , Humans , Immunologic Memory , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology
11.
Curr Opin Organ Transplant ; 23(1): 1-7, 2018 02.
Article in English | MEDLINE | ID: mdl-29210727

ABSTRACT

PURPOSE OF REVIEW: The main objective of this review is to briefly highlight how we gradually came to understand regulatory T cells (Tregs) and forkhead box p3 (FoxP3) biology, including their function and regulation. We will also discuss how this knowledge is being translated into the clinical setting and the significant challenges that need to be overcome. RECENT FINDINGS: CD4FoxP3 Tregs are key players in immune regulation. Their deficiency and dysfunction have been implicated in the pathogenesis of many autoimmune diseases. This has led towards extensive work across the years to figure out the biology and suppressive mechanisms of these cells. Furthermore, Tregs' ability to suppress immune responses makes the idea of their utilization in adoptive immunotherapy appealing. Work has been underway to establish ideal methods to integrate Tregs into the management of autoimmune diseases and alloimmunity, either by treatment with IL-2 or infusion of ex-vivo expanded Tregs. Despite Tregs' scarcity and increased tendency for Activation-induced cell death, many groups have developed effective methods to expand them ex vivo. SUMMARY: Although clinical trials are ongoing to test the safety and efficacy of regulatory cells in transplant recipients, it is vital to continue exploring the cellular and molecular mechanisms that control their stability and homeostasis.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Immunologic Factors/immunology , Immunotherapy , T-Lymphocytes, Regulatory/immunology , Animals , Humans
14.
Curr Opin Nephrol Hypertens ; 26(6): 509-515, 2017 11.
Article in English | MEDLINE | ID: mdl-28857783

ABSTRACT

PURPOSE OF REVIEW: Improved long-term kidney allograft survival remains a critical goal in transplantation; the achievement of this, however, is highly dependent on the identification of biomarkers that can either predict or allow advance detection of patients at risk of allograft injury. The present review outlines the commonly used biomarkers in kidney transplantation, while also highlighting those currently under investigation, discussing their advantages and limitations. RECENT FINDINGS: Most of the approved biomarkers currently used in kidney transplantation capture antigen recognition or alloantibody production. However, tremendous progress has recently been made in the development of markers of other signaling pathways pertinent to the alloimmune response. Microarray gene sets that predict rejection or poor prognostic phenotypes have been identified in kidney biopsies (the 'molecular microscope diagnostic system' and the 'genomics of chronic allograft rejection' scores), peripheral blood (the 'kidney solid organ response test'), and urine (the '3-genes signature'). Strategies targeting serial measurements of urinary chemokines such as CXCL9 and CXCL10 also appear promising. SUMMARY: Although the range of biomarkers in current use is limited, there are many assays in the development and validation pipeline that appear promising but that have yet to reach mainstream clinical transplantation. The 'ideal biomarker' may eventually transpire to be the combination of several assays.


Subject(s)
Biomarkers/metabolism , Graft Rejection/diagnosis , Graft Rejection/metabolism , Isoantibodies/blood , Kidney Transplantation , Chemokine CXCL10/urine , Chemokine CXCL9/urine , Gene Expression , Genomics , Graft Rejection/genetics , Humans , Kidney Transplantation/adverse effects
15.
Stem Cells ; 33(6): 1892-901, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25753288

ABSTRACT

The immunomodulatory capacity of mesenchymal stem cells (MSCs) is critical for their use in therapeutic applications. MSC response to specific inflammatory cues allows them to switch between a proinflammatory (MSC1) or anti-inflammatory (MSC2) phenotype. Regulatory mechanisms controlling this switch remain to be defined. One characteristic feature of MSC2 is their ability to respond to IFNγ with induction of indoleamine 2,3-dioxygenase (IDO), representing the key immunoregulatory molecule released by human MSC. Here, we show that STAT1 and PI3Kα pathways interplay regulates IFNγ-induced IDO production in MSC. Chemical phosphoinositide 3-kinase (PI3K) pan-inhibition, PI3Kα-specific inhibition or shRNA knockdown diminished IFNγ-induced IDO production. This effect involved PI3Kα-mediated upregulation of STAT1 protein levels and phosphorylation at Ser727. Overexpression of STAT1 or of a constitutively active PI3Kα mutant failed to induce basal IDO production, but shifted MSC into an MSC2-like phenotype by strongly enhancing IDO production in response to IFNγ as compared to controls. STAT1 overexpression strongly enhanced MSC-mediated T-cell suppression. The same effect could be induced using short-term pretreatment of MSC with a chemical inhibitor of the counter player of PI3K, phosphatase and tensin homolog. Finally, downregulation of STAT1 abrogated the immunosuppressive capacity of MSC. Our results for the first time identify critical upstream signals for the induced production of IDO in MSCs that could be manipulated therapeutically to enhance their immunosuppressive phenotype.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Phosphatidylinositol 3-Kinases/metabolism , STAT1 Transcription Factor/metabolism , Class I Phosphatidylinositol 3-Kinases , Down-Regulation , Humans , Interferon-gamma/metabolism , STAT3 Transcription Factor/metabolism , Up-Regulation
18.
J Immunol ; 191(12): 5785-91, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24319282

ABSTRACT

Calcineurin inhibitors (CNIs) revolutionized the field of organ transplantation and remain the standard of care 40 years after the discovery of cyclosporine. The early impressive results of cyclosporine in kidney transplant recipients led to its subsequent use in other organ transplant recipients and for treatment of a variety of autoimmune diseases as well. In this review, we examine the discovery of CNIs, their mechanism of action, preclinical and clinical studies with CNIs, and the usage of CNIs in nontransplant recipients. We review the mechanisms of renal toxicity associated with CNIs and the recent efforts to avoid or reduce usage of these drugs. Although minimization strategies are possible, safe, and of potential long-term benefit, complete avoidance of CNIs has proven to be more challenging than initially thought.


Subject(s)
Calcineurin Inhibitors , Cyclosporine/history , Immunosuppressive Agents/history , Animals , Autoimmune Diseases/drug therapy , Calcineurin/physiology , Clinical Trials as Topic , Cyclosporine/adverse effects , Cyclosporine/chemistry , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Diabetes Mellitus, Type 2/chemically induced , Drug Evaluation, Preclinical , Drug Therapy, Combination , Forecasting , Graft Rejection/prevention & control , History, 20th Century , History, 21st Century , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Kidney Diseases/immunology , Kidney Transplantation/history , Lymphocyte Activation/drug effects , Meta-Analysis as Topic , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus/adverse effects , Tacrolimus/chemistry , Tacrolimus/history , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
19.
J Immunol ; 191(5): 2319-27, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913965

ABSTRACT

Regulatory T cells (Tregs) play a pivotal role in the maintenance of immune tolerance and hold great promise as cell therapy for a variety of immune-mediated diseases. However, the cellular mechanisms that regulate Treg maintenance and homeostasis have yet to be fully explored. Although Tregs express granzyme-B (GrB) to suppress effector T cells via direct killing, the mechanisms by which they protect themselves from GrB-mediated self-inflicted damage are unknown. To our knowledge, we show for the first time that both induced Tregs and natural Tregs (nTregs) increase their intracellular expression of GrB and its endogenous inhibitor, serine protease inhibitor 6 (Spi6) upon activation. Subcellular fractionation and measurement of GrB activity in the cytoplasm of Tregs show that activated Spi6(-/-) Tregs had significantly higher cytoplasmic GrB activity. We observed an increase in GrB-mediated apoptosis in Spi6(-/-) nTregs and impaired suppression of alloreactive T cells in vitro. Spi6(-/-) Tregs were rescued from apoptosis by the addition of a GrB inhibitor (Z-AAD-CMK) in vitro. Furthermore, adoptive transfer experiments showed that Spi6(-/-) nTregs were less effective than wild type nTregs in suppressing graft-versus-host disease because of their impaired survival, as shown in our in vivo bioluminescence imaging. Finally, Spi6-deficient recipients rejected MHC class II-mismatch heart allografts at a much faster rate and showed a higher rate of apoptosis among Tregs, as compared with wild type recipients. To our knowledge, our data demonstrate, for the first time, a novel role for Spi6 in Treg homeostasis by protecting activated Tregs from GrB-mediated injury. These data could have significant clinical implications for Treg-based therapy in immune-mediated diseases.


Subject(s)
Granzymes/immunology , Homeostasis/immunology , Membrane Proteins/immunology , Serine Endopeptidases/immunology , Serpins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/immunology , Flow Cytometry , Graft vs Host Disease/immunology , Granzymes/metabolism , Heart Transplantation/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Serine Endopeptidases/metabolism , Serpins/metabolism , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL