Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 832488, 2022.
Article in English | MEDLINE | ID: mdl-35401566

ABSTRACT

Introduction: Kidney transplants fail more often in Black than in non-Black (White, non-Black Hispanic, and Asian) recipients. We used the estimated physicochemical immunogenicity for polymorphic amino acids of donor/recipient HLAs to select weakly immunogenic kidney transplants for Black vs. White or non-Black patients. Methods: OPTN data for 65,040 donor/recipient pairs over a 20-year period were used to calculate the individual physicochemical immunogenicity by hydrophobic, electrostatic and amino acid mismatch scores (HMS, EMS, AMS) and graft-survival outcomes for Black vs. White or vs. non-Black recipients, using Kaplan-Meier survival and Cox regression analyses. Simulations for re-matching recipients with donors were based on race-adjusted HMS thresholds with clinically achievable allocations. Results: The retrospective median kidney graft survival was 12.0 years in Black vs. 18.6 years in White (6.6-year difference; p>0.001) and 18.4 years in non-Black (6.4-year difference; p>0.01) recipients. Only 0.7% of Blacks received transplants matched at HLA-A/B/DR/DQ (HMS=0) vs. 8.1% in Whites (p<0.001). Among fully matched Blacks (HMS=0), graft survival was 16.1-years and in well-matched Blacks (HMS ≤ 3.0) it was 14.0-years. Whites had 21.6-years survival at HMS ≤ 3.0 and 18.7-years at HMS ≤ 7.0 whereas non-Blacks had 22.0-year at HMS ≤ 3.0 and 18.7-year at HMS ≤ 7.0, confirming that higher HMS thresholds produced excellent survival. Simulation of ABO-compatible donor-recipient pairs using race-adjusted HMS thresholds identified weakly immunogenic matches at HMS=0 for 6.1% Blacks and 18.0% at HMS ≤ 3.0. Despite prioritizing Black patients, non-Black patients could be matched at the same level as in current allocation (47.0% vs 56.5%, at HMS ≤ 7.0). Conclusions: Race-adjusted HMS (EMS, AMS)-based allocation increased the number of weakly immunogenic donors for Black patients, while still providing excellent options for non-Black recipients.


Subject(s)
Black or African American , Kidney Transplantation , Graft Survival , HLA Antigens , Humans , Kidney Transplantation/adverse effects , Retrospective Studies , Tissue Donors
2.
J Clin Med ; 10(4)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33562074

ABSTRACT

A growing body of evidence suggests that postural orthostatic tachycardia syndrome (POTS) may be an autoimmune disorder. We have reported in a previous manuscript that 89% of POTS patients (n = 55) had elevations in G-protein-coupled adrenergic A1 receptor autoantibodies and 53% had elevations in muscarinic acetylcholine M4 receptor autoantibodies, as assessed by ELISA. Patients with autoimmune disorders have been reported with a variety of elevated cytokines and cytokines (such as rheumatoid arthritis); thus, we evaluated a limited number of cytokines/chemokines in POTS patients with elevated adrenergic and muscarinic receptor autoantibodies. We utilized the plasma of 34 patients from a previous study; all of the patients (100%) had autoantibodies against the A1 adrenergic receptor and 55.9% (19/34) had autoantibodies against the M4 muscarinic acetylcholine receptor. In particular, the plasma cytokine/chemokine levels were measured as biomarkers of inflammation by Quantibody® technology (Raybiotech, Peachtree Corners, GA, USA). We also evaluated the platelet dense granule numbers, as these patients frequently complain of symptoms related to platelet dysfunction. Patients were predominantly young females who displayed a multitude of co-morbidities but generally reported viral-like symptoms preceding episodes of syncope. Eighty five percent (29/34) had platelet storage pool deficiency. Patients had elevations in five of ten cytokine/chemokines biomarkers (IL1ß, IL21, TNFα, INFγ, and CD30), whereas two biomarkers had decreased levels (CD40L and RANTES). Our observations demonstrate that POTS patients known to have autoantibodies against the G-protein-coupled adrenergic A1 receptor have abnormal plasma concentrations of inflammatory cytokines.

3.
Front Immunol ; 11: 580752, 2020.
Article in English | MEDLINE | ID: mdl-33193383

ABSTRACT

We evaluated the impact of human leukocyte antigen (HLA) disparity (immunogenicity; IM) on long-term kidney allograft survival. The IM was quantified based on physicochemical properties of the polymorphic linear donor/recipient HLA amino acids (the Cambridge algorithm) as a hydrophobic, electrostatic, amino acid mismatch scores (HMS\AMS\EMS) or eplet mismatch (EpMM) load. High-resolution HLA-A/B/DRB1/DQB1 types were imputed to calculate HMS for primary/re-transplant recipients of deceased donor transplants. The multiple Cox regression showed the association of HMS with graft survival and other confounders. The HMS integer 0-10 scale showed the most survival benefit between HMS 0 and 3. The Kaplan-Meier analysis showed that: the HMS=0 group had 18.1-year median graft survival, a 5-year benefit over HMS>0 group; HMS ≤ 3.0 had 16.7-year graft survival, a 3.8-year better than HMS>3.0 group; and, HMS ≤ 7.8 had 14.3-year grafts survival, a 1.8-year improvement over HMS>7.8 group. Stratification based on EMS, AMS or EpMM produced similar results. Additionally, the importance of HLA-DR with/without -DQ IM for graft survival was shown. In our simulation of 1,000 random donor/recipient pairs, 75% with HMS>3.0 were re-matched into HMS ≤ 3.0 and the remaining 25% into HMS≥7.8: after re-matching, the 13.5 years graft survival would increase to 16.3 years. This approach matches donors to recipients with low/medium IM donors thus preventing transplants with high IM donors.


Subject(s)
Graft Rejection/immunology , HLA-A Antigens/genetics , HLA-B Antigens/genetics , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Kidney Transplantation , Adolescent , Adult , Aged , Allografts , Amino Acids/chemistry , Amino Acids/genetics , Female , Genetic Loci , Graft Survival , HLA-A Antigens/metabolism , HLA-B Antigens/metabolism , HLA-DQ Antigens/metabolism , HLA-DR Antigens/metabolism , Histocompatibility Testing , Humans , Hydrophobic and Hydrophilic Interactions , Male , Middle Aged , Resource Allocation , Survival Analysis , Tissue Donors , Transplant Recipients , Young Adult
4.
Transpl Int ; 32(8): 839-853, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30848501

ABSTRACT

We examined what happened during a 6-year period to 1121 end-stage renal disease patients who registered with their willing/incompatible living donors for kidney exchanges with the Alliance for Paired Donation (APD). Of all patients, 65% were transplanted: 37% in kidney paired donation (APD-KPD, APD-other-KPD); 10% with compatible live donors (APD-LD); and 18% with deceased donors (APD-DD). The remaining patients were withdrawn (sick/died/others; 15%), or were still waiting (20%). For those patients with a cPRA 0-94%, 72% received a transplant. In contrast, only 49% of very highly sensitized (VHS; cPRA 95-100%) were transplanted. Of the VHS patients, 50% were transplanted by KPD/APD-LD while 50% benefited through prioritization of deceased donors in the modified kidney allocation system (KAS introduced in 2014). All APD transplanted groups had similar death-censored 4-year graft survivals as their relevant Organ Procurement and Transplantation Network (OPTN) groups. It is noteworthy that VHS graft and patient survival results were comparable to less sensitized and nonsensitized patients. All patients should be encouraged to search for compatible donors through different options. Expanding the donor pool through KPD and the new KAS of the OPTN increases the likelihood of transplantation for VHS patients.


Subject(s)
Kidney Failure, Chronic/surgery , Kidney Transplantation/methods , Tissue and Organ Procurement/methods , Adult , Algorithms , Databases, Factual , Family Health , Female , Graft Survival , Humans , Living Donors , Male , Middle Aged , Retrospective Studies , Treatment Outcome , United States
5.
Diabetologia ; 58(6): 1309-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25794782

ABSTRACT

AIMS/HYPOTHESIS: T cells play a major role in the pathogenesis of type 1 diabetes, and there is great interest in developing curative immunotherapies targeting these cells. In this study, a monoclonal antibody (mAb) targeting the T cell receptor ß-chain (TCRß) was investigated for its ability to prevent and reverse disease in mouse models of diabetes. METHODS: RIP-OVA(hi) (C57BL/6-Tg(Ins2-OVA)59Wehi/WehiJ) mice adoptively transferred with ovalbumin-specific T cells (an induced model of diabetes) and NOD mice (a spontaneous model of diabetes) were used to test anti-TCRß mAb therapy as a means of preventing and reversing type 1 diabetes. RESULTS: A single dose of anti-TCRß completely prevented disease in RIP-OVA(hi) mice without inducing the release of inflammatory cytokines. Transient anti-TCRß therapy prevented diabetes in 90% of NOD mice and reversed the disease after its onset in 73% of NOD mice. Long after the remission of type 1 diabetes, the anti-TCRß treated mice were able to reject BALB/c skin allografts with normal kinetics while maintaining normoglycaemia. Treatment did not cause significant reductions in lymphocyte numbers in the spleen or pancreatic lymph nodes, but did result in a decreased percentage of chemokine receptor 9 (CCR9) positive, CD8(+) T cells. Notably, anti-TCRß therapy increased the expression of programmed death 1 (PD-1) on the surface of the T cells; PD-1 expression is important for maintaining anti-TCRß-induced self-tolerance, as type 1 diabetes recurs in mice following a blockade of PD-1 signalling. CONCLUSIONS/INTERPRETATION: Anti-TCRß mAb is a safe and effective immunotherapy that results in reduced numbers of CCR9(+) T cells, an increased expression of PD-1 on T cells and the restoration of self-tolerance in NOD mice.


Subject(s)
Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Programmed Cell Death 1 Receptor/metabolism , Allografts , Animals , Blood Glucose/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Female , Glucose Tolerance Test , Inflammation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, CCR/metabolism
6.
Transplantation ; 97(12): 1216-24, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24854475

ABSTRACT

BACKGROUND: Clinical application of islet transplantation to treat type 1 diabetes has been limited by islet allograft destruction by both allogeneic and autoimmune diabetogenic T-cell responses. The current study aims at determining whether an anti-T-cell receptor (TCR) monoclonal antibody (mAb) has potential as a novel and potent induction immunotherapy for islet transplantation. METHODS: We have investigated the therapeutic efficacy and mechanisms of action of anti-TCR therapy in four different murine models, which comprise either allo- or autoimmune responses alone or both together. RESULTS: T-cell response to islet allografts was potently abrogated by a brief treatment with an anti-TCRß mAb (clone H57-597), resulting in long-term survival of BALB/c islet allografts in streptozotocin-induced diabetic B6 mice. Moreover, transient anti-TCR treatment permanently prevented BALB/c skin allograft rejection on Rag1 B6 recipients that were reconstituted with Foxp3 cell-depleted B6 splenocytes, but did not impair the reconstituted cells' ability to reject the later transplanted C3H skin allografts (transplanted at 120 days after BALB/c skin grafting). Transient anti-TCR treatment was also able to completely prevent diabetes onset in NOD.SCID.γc mice that were transferred with lymphocytes from diabetic NOD mice. Next, transient anti-TCR treatment significantly prolonged the survival of transplanted BALB/c islets in overtly diabetic NOD mice, which comprise both allogeneic and autoimmune diabetogenic T-cell responses to the transplanted islets. CONCLUSIONS: Overall, anti-TCR mAb induced peripheral tolerance to specific alloantigens even in the absence of Foxp3-expressing natural regulatory T cells. These findings reveal the potential for using TCR-targeting mAbs as induction immunotherapy for islet transplantation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Graft Rejection/prevention & control , Graft Survival/drug effects , Islets of Langerhans Transplantation , Isoantigens/immunology , Receptors, Antigen, T-Cell/immunology , Transplantation Tolerance/drug effects , Adoptive Transfer , Allografts , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/immunology , Graft Rejection/blood , Graft Rejection/immunology , Islets of Langerhans Transplantation/adverse effects , Isoantigens/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Skin Transplantation/adverse effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation, Homologous
7.
PLoS One ; 9(1): e85882, 2014.
Article in English | MEDLINE | ID: mdl-24416451

ABSTRACT

Optimal T cell activation and expansion require binding of the common gamma-chain (γc) cytokine Interleukin-2 (IL-2) to its cognate receptor that in turn engages a γc/Janus tyrosine kinase (Jak)3 signaling pathway. Because of its restricted expression by antigen-activated T cells and its obligatory role in promoting their survival and proliferation, IL-2 has been considered as a selective therapeutic target for preventing T cell mediated diseases. However, in order to further explore IL-2 targeted therapy, it is critical to precisely understand its role during early events of T cell activation. In this study, we delineate the role of IL-2 and other γc cytokines in promoting the survival of CD4 and CD8 T cells during early phases of priming. Under IL-2 inhibitory conditions (by neutralizing anti-IL-2 mAbs), the survival of activated CD8⁺ T cells was reduced, whereas CD4⁺ T cells remained much more resistant. These results correlated with reduced Bcl-2 expression, and mitochondrial membrane potential in CD8⁺ T cells in comparison to CD4⁺ T cells. However, using transwell co-culture assays we have found that CD4⁺ T cells could rescue the survival of CD8⁺ T cells even under IL-2 deprived conditions via secretion of soluble factors. A cytokine screen performed on CD8⁺ T cells cultured alone revealed that IL-21, another γc cytokine, was capable of rescuing their survival under IL-2 deprivation. Indeed, blocking the IL-21 signaling pathway along with IL-2 neutralization resulted in significantly reduced survival of both CD4⁺ and CD8⁺ T cells. Taken together, we have shown that under IL-2 deprivation conditions, IL-21 may act as the major survival factor promoting T cell immune responses. Thus, investigation of IL-2 targeted therapies may need to be revisited to consider blockade of the IL-21 signaling pathways as an adjunct to provide more effective control of T cell immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cross-Priming/drug effects , Interleukin-2/pharmacology , Interleukins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Survival/drug effects , Coculture Techniques , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism
8.
PLoS One ; 8(7): e69397, 2013.
Article in English | MEDLINE | ID: mdl-23894468

ABSTRACT

T cells play a major role in allograft rejection, which occurs after T cell activation by the engagement of several functional molecules to form an immune synapse with alloantigen presenting cells. In this study, the immune synapse was targeted using mAbs directed to the TCR beta-chain (TCRß) and lymphocyte function-associated antigen-1 (LFA1) to induce long-term allograft survival. Evaluation of antigen-specific T cell responses was performed by adoptively transferring CFSE labeled transgenic OT-II cells into wild-type mice and providing OVA peptide by intravenous injection. Graft survival studies were performed in mice by transplanting BALB/c ear skins onto the flanks of C57BL/6 recipients. The anti-TCRß plus anti-LFA1 mAb combination (but not either mAb alone) abrogated antigen-specific T cell responses invitro and invivo. Transient combination therapy with these agents resulted in significantly prolonged skin allograft survival in mice (51±10 days; p<0.01) when compared to treatment with either anti-TCRß mAb (24±5 days) or anti-LFA1 mAb (19±3 days) alone or no treatment (10±1 days). When lymphoid tissues from these mice were analyzed at different times post-transplant, only those receiving the combination of anti-TCRß and anti-LFA1 mAbs demonstrated long-lasting reductions in total T cell numbers, cellular and humoral anti-donor responses, and expression of CD3 on the surface of T cells. These results demonstrate that transient anti-TCRß and anti-LFA1 mAb combination therapy abrogates antigen-reactive T cell responses with long-lasting effects that significantly prolong allograft survival.


Subject(s)
Graft Survival/immunology , T-Lymphocytes/immunology , Allografts/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Flow Cytometry , Graft Rejection/immunology , Graft Rejection/prevention & control , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/immunology , Skin Transplantation
9.
Transplantation ; 95(7): 919-27, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23545505

ABSTRACT

BACKGROUND: KRP203, a structural FTY720 analogue, has 5-fold greater selectivity for binding to sphingosine-1-phosphate receptor (S1PR) 1 (S1PR(1)) versus S1PR3 and 100-fold greater selectivity over S1PR(2) and S1PR(5). Although the immunoregulatory effects of FTY720 have been tested in clinical and experimental research, the therapeutic efficacy of KRP203 in allograft models remains elusive. In this study, we investigated the potential of KRP203 alone and in combination with intragraft injection of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) to induce islet allograft tolerance. METHODS: BALB/c (H-2(d)) mice received transplants of fresh C57BL/10 (H-2(b)) islet allografts under the kidney capsule and were treated for 7 days with 0.3, 1.0, or 3.0 mg/kg KRP203 alone or in combination with intragraft-infused Tregs. RESULTS: Untreated BALB/c mice acutely rejected C57BL/10 islet allografts at a mean survival time of 13.8 ± 2.7 days (n=5). A 7-day dosing of 0.3 or 1.0 mg/kg KRP203 produced long-term islet allograft survival (9200 days) in one of five and two of seven recipients, respectively. A 3 mg/kg KRP203 dose resulted in islet graft survival for more than 200 days in 5 of 12 recipients. Whereas recipients that received 500 allogeneic islets admixed with 5 x 10(5) - 7 x 10(5) Tregssurvived 83.6 ± 67.2 days, addition of transient 3 mg/kg KRP203 therapy induced prolonged drug-free graft survival (9200 days) in all recipients. CONCLUSIONS: A brief treatment with KRP203 significantly prolonged islet allograft survival, whereas additional intragraft delivery of Tregs induced tolerogenic effects selective to islet alloantigens.


Subject(s)
Adoptive Transfer , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Receptors, Lysosphingolipid/drug effects , Sulfhydryl Compounds/pharmacology , T-Lymphocytes, Regulatory/transplantation , Animals , Biomarkers/metabolism , Cells, Cultured , Combined Modality Therapy , Dose-Response Relationship, Drug , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Graft Rejection/immunology , Graft Rejection/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Islets of Langerhans Transplantation/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation Tolerance/drug effects
10.
J Immunol ; 190(7): 3153-62, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23427250

ABSTRACT

The molecular mechanism of the extrathymic generation of adaptive, or inducible, CD4(+)Foxp3(+) regulatory T cells (iTregs) remains incompletely defined. We show that exposure of splenic CD4(+)CD25(+)Foxp3(-) cells to IL-2, but not other common γ-chain cytokines, resulted in Stat5 phosphorylation and induced Foxp3 expression in ∼10% of the cells. Thus, IL-2/Stat5 signaling may be critical for Foxp3 induction in peripheral CD4(+)CD25(+)Foxp3(-) iTreg precursors. In this study, to further define the role of IL-2 in the formation of iTreg precursors as well as their subsequent Foxp3 expression, we designed a two-step iTreg differentiation model. During the initial "conditioning" step, CD4(+)CD25(-)Foxp3(-) naive T cells were activated by TCR stimulation. Inhibition of IL-2 signaling via Jak3-Stat5 was required during this step to generate CD4(+)CD25(+)Foxp3(-) cells containing iTreg precursors. During the subsequent Foxp3-induction step driven by cytokines, IL-2 was the most potent cytokine to induce Foxp3 expression in these iTreg precursors. This two-step method generated a large number of iTregs with relatively stable expression of Foxp3, which were able to prevent CD4(+)CD45RB(high) cell-mediated colitis in Rag1(-/-) mice. In consideration of this information, whereas initial inhibition of IL-2 signaling upon T cell priming generates iTreg precursors, subsequent activation of IL-2 signaling in these precursors induces the expression of Foxp3. These findings advance the understanding of iTreg differentiation and may facilitate the therapeutic use of iTregs in immune disorders.


Subject(s)
Cell Differentiation , Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Colitis/immunology , Colitis/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Knockout , Phosphorylation/drug effects , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/drug effects , Precursor Cells, T-Lymphoid/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/metabolism
11.
J Immunol ; 186(6): 3753-61, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21307294

ABSTRACT

Maraviroc (MVC), a specific antagonist of CCR5 expressed on macrophages and activated T cells, may modulate inflammation and may be useful in patients with HIV infection. In this study we used nonhuman primates to examine the effect and mechanism of MVC alone or in combination with cyclosporine (CsA) to prolong cardiac allograft survivals. In an established rhesus monkey cardiac allograft model, recipients treated with MVC plus CsA showed significantly prolonged survival of heart allografts (>240 d, p < 0.001). These in vivo results in the MVC/CsA group correlated with delayed alloantibody response and markedly decreased graft infiltration by CCR5(+), CD4(+), CD8(+), and CD68(+) cells (p < 0.05), as compared with other groups. Furthermore, grafts from the MVC/CsA group had elevated numbers of alternatively activated macrophages (AAMs) and the expression of peroxisome proliferator-activated receptor γ (PPARγ). Blockade of PPARγ abrogated the prolonged allograft survival (median survival time, 45 d) and the upregulated AAMs in MVC/CsA-treated recipients. In conclusion, MVC/CsA protects cardiac allograft in primates and this effect is associated with generating AAMs through activation of the PPARγ nuclear receptor.


Subject(s)
CCR5 Receptor Antagonists , Cyclohexanes/therapeutic use , Cyclosporine/therapeutic use , Graft Survival/immunology , Heart Transplantation/immunology , Macrophage Activation/immunology , Triazoles/therapeutic use , Animals , Binding, Competitive/drug effects , Binding, Competitive/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Drug Therapy, Combination , Graft Survival/drug effects , Lymphocyte Culture Test, Mixed , Macaca mulatta , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/immunology , Male , Maraviroc , PPAR gamma/biosynthesis , PPAR gamma/physiology , Protein Binding/drug effects , Protein Binding/immunology , Receptors, CCR5/physiology , Up-Regulation/drug effects , Up-Regulation/immunology
12.
Immunol Lett ; 132(1-2): 61-8, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20570629

ABSTRACT

The benefits of immunotherapy by regulatory T (Treg) cells are unpredictable partially due to the uncertainty of their suppressive mechanism. In fact, various suppressive mechanisms have been proposed but each remains controversial. To better understand Treg-mediated suppression, we have investigated factors which may influence the suppressive effects. In an in vitro suppression assay, over-expression of anti-apoptotic Bcl2 enhancing survival of conventional T responder cells (Tconvs) did not subvert Treg-mediated suppression. In contrast, enhancing activation of Tconvs by increasing the potency of calcium signals completely abrogated Treg-mediated suppression. While Tregs were incapable of suppressing already activated Tconvs, they prevented expression of activation markers on naïve Tconvs during activation, thereby indicating that Tregs mediate suppression through controlling early activation stage. Interestingly, IL-2 deprivation or TGF-beta, two suppressive mechanisms, did not effectively inhibit Tconv activation and proliferation when applied alone. In contrast, IL-2 deprivation combined with TGF-beta suppressed Tconv activation as potently as Tregs. More importantly, in the transwell system, that separates Tregs from Tconvs, TGF-beta contributed to Treg suppression under IL-2 depriving condition. In conclusion, these two suppressive mechanisms acting in concert may be necessary to effectively restrain the early activation of Tconvs.


Subject(s)
Immune Tolerance , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology , Animals , CD24 Antigen/metabolism , Cells, Cultured , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
13.
J Immunol ; 185(1): 71-8, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20498359

ABSTRACT

CD4(+)Foxp3(+) regulatory T (Treg) cells were shown to control all aspects of immune responses. How these Treg cells develop is not fully defined, especially in neonates during development of the immune system. We studied the induction of Treg cells from neonatal T cells with various TCR stimulatory conditions, because TCR stimulation is required for Treg cell generation. Independent of the types of TCR stimulus and without the addition of exogenous TGF-beta, up to 70% of neonatal CD4(+)Foxp3(-) T cells became CD4(+)Foxp3(+) Treg cells, whereas generally <10% of adult CD4(+)Foxp3(-) T cells became CD4(+)Foxp3(+) Treg cells under the same conditions. These neonatal Treg cells exert suppressive function and display relatively stable Foxp3 expression. Importantly, this ability of Treg cell generation gradually diminishes within 2 wk of birth. Consistent with in vitro findings, the in vivo i.p. injection of anti-CD3 mAb to stimulate T cells also resulted in a >3-fold increase in Treg cells in neonates but not in adults. Furthermore, neonatal or adult Foxp3(-) T cells were adoptively transferred into Rag1(-/-) mice. Twelve days later, the frequency of CD4(+)Foxp3(+) T cells converted from neonatal cells was 6-fold higher than that converted from adult cells. Taken together, neonatal CD4(+) T cells have an intrinsic "default" mechanism to become Treg cells in response to TCR stimulations. This finding provides intriguing implications about neonatal immunity, Treg cell generation, and tolerance establishment early in life.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/biosynthesis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Animals, Newborn , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , Cell Differentiation/genetics , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/immunology , Female , Forkhead Transcription Factors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Stability , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
14.
Immunol Lett ; 128(2): 116-23, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20064560

ABSTRACT

Multiple activation signals (including antigen, co-stimulation, and cytokines) during T-cell priming affect the subsequent generation of memory T cells, whose survival is maintained by IL-7 and IL-15. Since the IL-7 receptor is highly expressed not only on the surface of memory T cells but also on naïve T cells, we propose that early exposure to IL-7 during priming of naïve T cells may promote their survival, and thus enhances the generation of memory cells. To test this hypothesis, TCR transgenic OT-II CD4(+) T cells were stimulated in vitro with OVA(323-339) peptide presented by syngeneic antigen-presenting cells (APCs). IL-7 or an IL-7 like cytokine, thymic stromal lymphopoietin (TSLP), was added at the initial 2-day cultivation stage. We found that a short exposure to IL-7 or TSLP during priming did not affect activation, proliferation, and glucose uptake by CD4(+) T cells compared to controls when examined on culture day 6. However, the same 6-day cultures showed that IL-7 (but not TSLP) significantly decreased the frequency of apoptotic CD4(+) T cells compared to controls. More importantly, an adoptive transfer of the 6-day primed OT-II T cells into CD45.1(+) congenic mice demonstrated that IL-7 (but not TSLP) elevated by 3- and 4-fold the number of transferred CD4(+) T cells in spleen (p<0.05) and lymph nodes (p<0.05), respectively, compared to controls. Almost all transferred CD4(+) T populations displayed phenotypes of effector (CD44(+)CD62L(-)) or central (CD44(+)CD62L(+)) memory T cells. We thus conclude that exposure of CD4(+) T cells to IL-7 during priming results in an increased frequency of CD4(+) memory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Immunologic Memory/immunology , Interleukin-7/immunology , Lymphocyte Activation/immunology , Animals , Cells, Cultured , Mice , Mice, Congenic , Mice, Inbred C57BL , Thymic Stromal Lymphopoietin
15.
Transplantation ; 88(12): 1323-31, 2009 Dec 27.
Article in English | MEDLINE | ID: mdl-20029328

ABSTRACT

BACKGROUND: We evaluated the impact of reduced nephron mass on nephrotoxicity by cyclosporine A (CsA) and/or sirolimus (SRL). METHODS: Renal function was tested in salt-depleted rats bearing two kidneys (2K), one kidney, or half a kidney (1/2K) and treated for 7 or 28 days with CsA (5 mg/kg) and/or SRL (0.8 mg/kg). We also measured the expression of aquaporin-2, sodium/phosphate cotransporter (NaPi)-2, paracellin-1, and kidney injury molecule (KIM)-1 by real-time polymerase chain reaction. RESULTS: At 7 days in 2K, serum creatinine clearance (CrCl) was decreased only in CsA/SRL-treated group (P<0.05) compared with controls; in 1/2K, CrCl was decreased in all groups, but most dramatically in CsA/SRL group (P<0.05). Extended 28-day therapy worsened CrCl in all 1/2K groups (P<0.01). Although the expression of aquaporin-2, NaPi-2, and paracellin-1 mRNAs tended to increase in kidneys with a reduced nephron mass, NaPi-2 mRNA levels decreased in 1/2K rats exposed to CsA/SRL for 28 days (P<0.05). In contrast, low KIM-1 mRNA expression in control 2K rats increased fourfold in untreated 1/2K (P<0.05), and 50- to 200-fold in CsA/SRL-treated 1/2K (P=0.01). CONCLUSIONS: Nephrotoxicity is significantly worsened by reduced nephron mass, which correlates with increased expression of KIM-1 and inhibited expression of NaPi-2.


Subject(s)
Cyclosporine/toxicity , Kidney Diseases/pathology , Nephrons/pathology , Sirolimus/toxicity , Animals , Aquaporin 2/biosynthesis , Aquaporin 2/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Creatinine/metabolism , Cyclosporine/pharmacokinetics , Disease Models, Animal , Follow-Up Studies , Gene Expression Regulation/drug effects , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Nephrons/drug effects , Nephrons/metabolism , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Risk Factors , Sirolimus/pharmacokinetics , Sodium-Phosphate Cotransporter Proteins/biosynthesis , Sodium-Phosphate Cotransporter Proteins/genetics
16.
Arch Immunol Ther Exp (Warsz) ; 57(3): 199-204, 2009.
Article in English | MEDLINE | ID: mdl-19479206

ABSTRACT

The human immune system is a myriad of diverse cellular populations, each contributing to maintaining an effective and optimal immune response against infectious agents. It is important to maintain a "self-check" in the immune system so that responses do not go haywire, leading to the development of autoimmune diseases. Regulatory/suppressor T (Treg) cells are a specialized subpopulation of T cells that suppress the activation, expansion, and function of other T cells, thereby maintaining homeostasis through a fine balance between reactivity to foreign and self antigens. Tregs are characterized by surface expression of interleukin (IL)-2 receptor alpha chain (CD25) and intracellular expression of forkhead box protein P3 (FoxP3). There are at least two important functional populations of Treg cells, namely natural Treg (nTreg), which are continuously derived from the thymus, and induced Treg (iTreg), which are converted from naive T cells. The development and function of both nTreg and iTreg cells are regulated by several factors, such as antigen T-cell receptor, co-stimulatory receptors (i.e., cytotoxic T lymphocyte-associated antigen, or CTLA-4), and cytokines (IL-2, IL-10, and tumor growth factor-beta, or TGF-beta). In addition, the TGF-beta inhibitor ALK5, retinoid acid, and rapamycin influence the expansion of nTreg cells and the conversion of iTreg cells in vitro and in vivo. The heightening of Treg expansion may be harnessed to therapeutic methods for the treatment of autoimmune diseases and the induction of transplantation tolerance.


Subject(s)
Immunotherapy/methods , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Differentiation/immunology , Cytokines/immunology , Cytokines/metabolism , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
18.
Kidney Int ; 74(10): 1319-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18769369

ABSTRACT

Agonists of the type 1 sphingosine-1-phosphate (S1P) receptor inhibit lymphocyte migration, causing their sequestration in lymphoid tissue. The S1P agonist FTY720 prolongs the survival of organ allografts and blocks T-cell mediated autoimmune diseases in experimental models; however, it is a non-selective agonist of four of the five S1P receptors. In this study female MRL/lpr mice, which develop an aggressive form of spontaneous autoimmune kidney disease, were treated with a more selective agonist of the type 1 receptor (KRP-203) or vehicle at 12 or 16 weeks of age. Eighty percent of the mice treated at 12 weeks, before the onset of visible disease, survived to the 24 weeks end point with decreased tubulointerstitial disease and significantly fewer infiltrating CD4(+) and CD8(+) T-cells. Only half of the control vehicle-treated mice survived. All of the mice treated at 16 weeks survived with reduced proteinuria. Mice in both groups had significant reductions in circulating lymphocytes. Mice receiving KRP-203 for 8-12 weeks had significant reductions in T-cells and consequently less adenopathy. Ex vivo treatment of lymphocytes from MRL/lpr mice with KRP-203 enhanced their apoptosis. Our study indicates that KRP-203 attenuates kidney injury in MRL/lpr mice, in part, by reducing T-cell infiltrates.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Lupus Nephritis/drug therapy , Receptors, Lysosphingolipid/agonists , Sulfhydryl Compounds/pharmacology , Animals , Apoptosis , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Lupus Nephritis/mortality , Lymphocytes , Mice , Mice, Inbred MRL lpr , Sulfhydryl Compounds/therapeutic use , Survival Rate
19.
Transplantation ; 85(10): 1372-7, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18497672

ABSTRACT

Maintaining T cell homeostasis is critical for normal immune response. Three sequential signals activate T cells, with signal 3 delivered by multiple cytokines that regulate cell proliferation, differentiation, and survival/death. Cytokines binding to their receptors engages two key molecular families, namely, Janus tyrosine kinases (Jaks) and signal transducers and activators of transcription (Stats). Among Stats, Stat3 is involved in the generation of T helper 17 (Th17) cells, regulation of dendritic cells, and acute inflammatory response. These aspects of Stat3 function are important for transplantation. We discuss Stat3's role in innate and adaptive immunity as well as its potential for therapeutic intervention.


Subject(s)
Cytokines/physiology , Immune Tolerance , STAT3 Transcription Factor/physiology , Homeostasis , Humans , Job Syndrome/genetics , Mutation , Neoplasms/physiopathology , STAT3 Transcription Factor/genetics , Signal Transduction , Transplantation Immunology
20.
Arch Immunol Ther Exp (Warsz) ; 55(4): 231-45, 2007.
Article in English | MEDLINE | ID: mdl-17659375

ABSTRACT

Regulation of T cell homeostasis is critical for maintaining normal immune function. An imbalance in T cell proliferation can result in disorders ranging from cancer and autoimmunity to immunodeficiencies. Full activation of T cells requires three sequential signals, where signal 3, which is delivered by multiple cytokines, regulates proliferation, differentiation, and survival/death. Signaling from cytokines through their receptors is primarily delivered by two molecular families, namely Janus tyrosine kinases (JAKs) and signal transducers and activators of transcription (STATs). Invaluable knowledge about JAKs and STATs has arisen from studies of mice made genetically deficient in these molecules, analyses of tumor models, and studies of expression patterns by proteomics/genomics, which all have begun to define the role of JAKs and STATs in survival versus apoptosis. These findings also have suggested ways in which JAKs and STATs may be manipulated for therapeutic intervention in lymphoid-derived diseases. This review seeks to focus on the role of JAK tyrosine kinases and STAT transcription factors in mediating the lymphocyte life cycle and how they might be manipulated for therapeutic applications.


Subject(s)
Cytokines/metabolism , Janus Kinases/metabolism , Lymphocyte Activation , STAT Transcription Factors/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Survival , Homeostasis , Humans , Immune Tolerance , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Janus Kinases/chemistry , Janus Kinases/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Neoplasms/immunology , Neoplasms/metabolism , Protein Conformation , STAT Transcription Factors/chemistry , STAT Transcription Factors/genetics , Signal Transduction/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...