Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 253
Filter
1.
Horm Metab Res ; 53(3): 204-206, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33652492

ABSTRACT

Currently, we are experiencing a true pandemic of a communicable disease by the virus SARS-CoV-2 holding the whole world firmly in its grasp. Amazingly and unfortunately, this virus uses a metabolic and endocrine pathway via ACE2 to enter our cells causing damage and disease. Our international research training programme funded by the German Research Foundation has a clear mission to train the best students wherever they may come from to learn to tackle the enormous challenges of diabetes and its complications for our society. A modern training programme in diabetes and metabolism does not only involve a thorough understanding of classical physiology, biology and clinical diabetology but has to bring together an interdisciplinary team. With the arrival of the coronavirus pandemic, this prestigious and unique metabolic training programme is facing new challenges but also new opportunities. The consortium of the training programme has recognized early on the need for a guidance and for practical recommendations to cope with the COVID-19 pandemic for the community of patients with metabolic disease, obesity and diabetes. This involves the optimal management from surgical obesity programmes to medications and insulin replacement. We also established a global registry analyzing the dimension and role of metabolic disease including new onset diabetes potentially triggered by the virus. We have involved experts of infectious disease and virology to our faculty with this metabolic training programme to offer the full breadth and scope of expertise needed to meet these scientific challenges. We have all learned that this pandemic does not respect or heed any national borders and that we have to work together as a global community. We believe that this transCampus metabolic training programme provides a prime example how an international team of established experts in the field of metabolism can work together with students from all over the world to address a new pandemic.


Subject(s)
COVID-19 , Diabetes Mellitus , Education, Medical, Continuing , Obesity , Pandemics , SARS-CoV-2 , COVID-19/epidemiology , COVID-19/therapy , Diabetes Mellitus/epidemiology , Diabetes Mellitus/therapy , Humans , Obesity/epidemiology , Obesity/therapy
2.
3.
Sci Rep ; 8(1): 2725, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426942

ABSTRACT

Galectin-1 (Gal-1) is required for the development of B cells in the bone marrow (BM), however very little is known about the contribution of Gal-1 to the development of B cell regulatory function. Here, we report an important role for Gal-1 in the induction of B cells regulatory function. Mice deficient of Gal-1 (Gal-1-/-) showed significant loss of Transitional-2 (T2) B cells, previously reported to include IL-10+ regulatory B cells. Gal-1-/- B cells stimulated in vitro via CD40 molecules have impaired IL-10 and Tim-1 expression, the latter reported to be required for IL-10 production in regulatory B cells, and increased TNF-α expression compared to wild type (WT) B cells. Unlike their WT counterparts, T2 and T1 Gal-1-/- B cells did not suppress TNF-α expression by CD4+ T cells activated in vitro with allogenic DCs (allo-DCs), nor were they suppressive in vivo, being unable to delay MHC-class I mismatched skin allograft rejection following adoptive transfer. Moreover, T cells stimulated with allo-DCs show an increase in their survival when co-cultured with Gal-1-/- T2 and MZ B cells compared to WT T2 and MZ B cells. Collectively, these data suggest that Gal-1 contributes to the induction of B cells regulatory function.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Dendritic Cells/immunology , Galectin 1/physiology , Graft Survival , Lymphocyte Activation , Allografts , Animals , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/pathology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Skin Transplantation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
4.
Clin Exp Immunol ; 189(2): 197-210, 2017 08.
Article in English | MEDLINE | ID: mdl-28422316

ABSTRACT

The concept of regulatory T cell (Treg ) therapy in transplantation is now a reality. Significant advances in science and technology have enabled us to isolate human Tregs , expand them to clinically relevant numbers and infuse them into human transplant recipients. With several Phase I/II trials under way investigating Treg safety and efficacy it is now more crucial than ever to understand their complex biology. However, our journey is by no means complete; results from these trials will undoubtedly provoke both further knowledge and enquiry which, alongside evolving science, will continue to drive the optimization of Treg therapy in the pursuit of transplantation tolerance. In this review we will summarize current knowledge of Treg biology, explore novel technologies in the setting of Treg immunotherapy and address key prerequisites surrounding the clinical application of Tregs in transplantation.


Subject(s)
Adaptive Immunity , Graft Rejection/drug therapy , Immunotherapy/methods , Organ Transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance , Animals , Clinical Trials as Topic , Cryopreservation , Humans , Immunosuppressive Agents/therapeutic use , Mice
5.
Clin Exp Immunol ; 188(2): 219-225, 2017 05.
Article in English | MEDLINE | ID: mdl-28120329

ABSTRACT

Anti-apoptotic genes, including those of the Bcl-2 family, have been shown to have dual functionality inasmuch as they inhibit cell death but also regulate inflammation. Several anti-apoptotic molecules have been associated with endothelial cell (EC) survival following transplantation; however, their exact role has yet to be elucidated in respect to controlling inflammation. In this study we created mice expressing murine A1 (Bfl-1), a Bcl-2 family member, under the control of the human intercellular adhesion molecule 2 (ICAM-2) promoter. Constitutive expression of A1 in murine vascular ECs conferred protection from cell death induced by the proinflammatory cytokine tumour necrosis factor (TNF)-α. Importantly, in a mouse model of heart allograft transplantation, expression of A1 in vascular endothelium increased survival in the absence of CD8+ T cells. Better graft outcome in mice receiving an A1 transgenic heart correlated with a reduced immune infiltration, which may be related to increased EC survival and reduced expression of adhesion molecules on ECs. In conclusion, constitutive expression of the anti-apoptotic molecule Bfl1 (A1) in murine vascular ECs leads to prolonged allograft survival due to modifying inflammation.


Subject(s)
Endothelial Cells/metabolism , Gene Expression , Heart Transplantation , Minor Histocompatibility Antigens/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Transplantation Tolerance , Animals , Antigens, CD/genetics , Apoptosis , CD8-Positive T-Lymphocytes , Cell Adhesion Molecules/genetics , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Graft Survival , Humans , Inflammation , Mice , Promoter Regions, Genetic , Transplantation, Homologous , Tumor Necrosis Factor-alpha/pharmacology
6.
Am J Transplant ; 17(4): 931-943, 2017 04.
Article in English | MEDLINE | ID: mdl-28027623

ABSTRACT

Regulatory T cell (Treg) therapy using recipient-derived Tregs expanded ex vivo is currently being investigated clinically by us and others as a means of reducing allograft rejection following organ transplantation. Data from animal models has demonstrated that adoptive transfer of allospecific Tregs offers greater protection from graft rejection compared to polyclonal Tregs. Chimeric antigen receptors (CAR) are clinically translatable synthetic fusion proteins that can redirect the specificity of T cells toward designated antigens. We used CAR technology to redirect human polyclonal Tregs toward donor-MHC class I molecules, which are ubiquitously expressed in allografts. Two novel HLA-A2-specific CARs were engineered: one comprising a CD28-CD3ζ signaling domain (CAR) and one lacking an intracellular signaling domain (ΔCAR). CAR Tregs were specifically activated and significantly more suppressive than polyclonal or ΔCAR Tregs in the presence of HLA-A2, without eliciting cytotoxic activity. Furthermore, CAR and ΔCAR Tregs preferentially transmigrated across HLA-A2-expressing endothelial cell monolayers. In a human skin xenograft transplant model, adoptive transfer of CAR Tregs alleviated the alloimmune-mediated skin injury caused by transferring allogeneic peripheral blood mononuclear cells more effectively than polyclonal Tregs. Our results demonstrated that the use of CAR technology is a clinically applicable refinement of Treg therapy for organ transplantation.


Subject(s)
Graft Rejection/prevention & control , HLA-A2 Antigen/immunology , Receptors, Antigen/immunology , Skin Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , Graft Rejection/etiology , Graft Survival/immunology , Heterografts , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred BALB C , Transplantation Tolerance/immunology
7.
Am J Transplant ; 17(1): 60-68, 2017 01.
Article in English | MEDLINE | ID: mdl-27495898

ABSTRACT

Understanding the evolution of the direct and indirect pathways of allorecognition following tissue transplantation is essential in the design of tolerance-promoting protocols. On the basis that donor bone marrow-derived antigen-presenting cells are eliminated within days of transplantation, it has been argued that the indirect response represents the major threat to long-term transplant survival, and is consequently the key target for regulation. However, the detection of MHC transfer between cells, and particularly the capture of MHC:peptide complexes by dendritic cells (DCs), led us to propose a third, semidirect, pathway of MHC allorecognition. Persistence of this pathway would lead to sustained activation of direct-pathway T cells, arguably persisting for the life of the transplant. In this study, we focused on the contribution of acquired MHC-class I on recipient DCs during the life span of a skin graft. We observed that MHC-class I acquisition by recipient DCs occurs for at least 1 month following transplantation and may be the main source of alloantigen that drives CD8+ cytotoxic T cell responses. In addition, acquired MHC-class I:peptide complexes stimulate T cell responses in vivo, further emphasizing the need to regulate both pathways to induce indefinite survival of the graft.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft Rejection/immunology , Immunity, Cellular/immunology , Isoantigens/immunology , Peptide Fragments/immunology , Skin Transplantation/adverse effects , Allografts , Animals , Antigen Presentation/immunology , Graft Rejection/pathology , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL , Tissue Donors , Transplant Recipients
8.
Am J Transplant ; 16(12): 3443-3457, 2016 12.
Article in English | MEDLINE | ID: mdl-27328267

ABSTRACT

We and others have previously described signatures of tolerance in kidney transplantation showing the differential expression of B cell-related genes and the relative expansions of B cell subsets. However, in all of these studies, the index group-namely, the tolerant recipients-were not receiving immunosuppression (IS) treatment, unlike the rest of the comparator groups. We aimed to assess the confounding effect of these regimens and develop a novel IS-independent signature of tolerance. Analyzing gene expression in three independent kidney transplant patient cohorts (232 recipients and 14 tolerant patients), we have established that the expression of the previously reported signature was biased by IS regimens, which also influenced transitional B cells. We have defined and validated a new gene expression signature that is independent of drug effects and also differentiates tolerant patients from healthy controls (cross-validated area under the receiver operating characteristic curve [AUC] = 0.81). In a prospective cohort, we have demonstrated that the new signature remained stable before and after steroid withdrawal. In addition, we report on a validated and highly accurate gene expression signature that can be reliably used to identify patients suitable for IS reduction (approximately 12% of stable patients), irrespective of the IS drugs they are receiving. Only a similar approach will make the conduct of pilot clinical trials for IS minimization safe and hence allow critical improvements in kidney posttransplant management.


Subject(s)
Biomarkers/metabolism , Graft Rejection/diagnosis , Graft Survival/immunology , Immune Tolerance/immunology , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/adverse effects , Adult , Aged , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/drug therapy , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Survival/drug effects , Humans , Immune Tolerance/drug effects , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/surgery , Kidney Function Tests , Male , Middle Aged , Prognosis , Prospective Studies , Risk Factors
9.
Immunol Lett ; 171: 36-49, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26835593

ABSTRACT

The gut harbors a complex community of over 100 trillion microbial cells known to exist in symbiotic harmony with the host influencing human physiology, metabolism, nutrition and immune function. It is now widely accepted that perturbations of this close partnership results in the pathogenesis of several major diseases with increasing evidence highlighting their role outside of the intestinal tract. The intimate proximity and circulatory loop of the liver and the gut has attracted significant attention regarding the role of the microbiota in the development and progression of liver disease. Here we give an overview of the interaction between the microbiota and the immune system and focus on their convincing role in both the propagation and treatment of liver disease.


Subject(s)
Immune System/microbiology , Immunity , Liver Diseases/microbiology , Liver/microbiology , Microbiota/immunology , Animals , Humans , Liver/immunology , Liver Diseases/immunology , Symbiosis
10.
Methods Mol Biol ; 1371: 177-96, 2016.
Article in English | MEDLINE | ID: mdl-26530801

ABSTRACT

Animal models have been instrumental in our understanding of the mechanisms of rejection and the testing of novel treatment options in the context of transplantation. We have now entered an exciting era with research on humanized mice driving advances in translational studies and in our understanding of the function of human cells in response to pathogens and cancer as well as the recognition of human allogeneic tissues in vivo. In this chapter we provide a historical overview of humanized mouse models of transplantation to date, outlining the distinct strains and share our experiences in the study of human transplantation immunology.


Subject(s)
Mice, Transgenic , Models, Animal , Transplantation Immunology , Transplantation , Animals , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Mice , Mice, Knockout , Transplantation/adverse effects , Transplantation/methods
11.
Sci Rep ; 5: 11554, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26109230

ABSTRACT

B cells have been reported to promote graft rejection through alloantibody production. However, there is growing evidence that B cells can contribute to the maintenance of tolerance. Here, we used a mouse model of MHC-class I mismatched skin transplantation to investigate the contribution of B cells to graft survival. We demonstrate that adoptive transfer of B cells prolongs skin graft survival but only when the B cells were isolated from mice housed in low sterility "conventional" (CV) facilities and not from mice housed in pathogen free facilities (SPF). However, prolongation of skin graft survival was lost when B cells were isolated from IL-10 deficient mice housed in CV facilities. The suppressive function of B cells isolated from mice housed in CV facilities correlated with an anti-inflammatory environment and with the presence of a different gut microflora compared to mice maintained in SPF facilities. Treatment of mice in the CV facility with antibiotics abrogated the regulatory capacity of B cells. Finally, we identified transitional B cells isolated from CV facilities as possessing the regulatory function. These findings demonstrate that B cells, and in particular transitional B cells, can promote prolongation of graft survival, a function dependent on licensing by gut microflora.


Subject(s)
B-Lymphocytes/immunology , Gastrointestinal Microbiome , Skin Transplantation , Adaptive Immunity , Adoptive Transfer , Animals , Anti-Bacterial Agents/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Graft Survival/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Immune Tolerance , Interleukin-10/deficiency , Interleukin-10/genetics , Lipopolysaccharides/toxicity , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , Spleen/pathology , Transplantation, Homologous
13.
Am J Transplant ; 13(11): 3010-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24102808

ABSTRACT

Regulatory T cell (Treg) therapy has the potential to induce transplantation tolerance so that immunosuppression and associated morbidity can be minimized. Alloantigen-reactive Tregs (arTregs) are more effective at preventing graft rejection than polyclonally expanded Tregs (PolyTregs) in murine models. We have developed a manufacturing process to expand human arTregs in short-term cultures using good manufacturing practice-compliant reagents. This process uses CD40L-activated allogeneic B cells to selectively expand arTregs followed by polyclonal restimulation to increase yield. Tregs expanded 100- to 1600-fold were highly alloantigen reactive and expressed the phenotype of stable Tregs. The alloantigen-expanded Tregs had a diverse TCR repertoire. They were more potent than PolyTregs in vitro and more effective at controlling allograft injuries in vivo in a humanized mouse model.


Subject(s)
Cell- and Tissue-Based Therapy , Graft Rejection/prevention & control , Immune Tolerance/immunology , Isoantigens/immunology , Skin Transplantation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Animals , Flow Cytometry , Graft Rejection/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Transplantation Tolerance
14.
Clin Exp Immunol ; 172(2): 158-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23574313

ABSTRACT

Transplantation is a successful treatment for end-stage organ failure. Despite improvements in short-term outcome, long-term survival remains suboptimal because of the morbidity and mortality associated with long-term use of immunosuppression. There is, therefore, a pressing need to devise protocols that induce tolerance in order to minimize or completely withdraw immunosuppression in transplant recipients. In this review we will discuss how regulatory T cells (T(regs)) came to be recognized as an attractive way to promote transplantation tolerance. We will summarize the preclinical data, supporting the importance of these cells in the induction and maintenance of immune tolerance and that provide the rationale for the isolation and expansion of these cells for cellular therapy. We will also describe the data from the first clinical trials, using T(regs) to inhibit graft-versus-host disease (GVHD) after haematopoietic stem cell transplantation and will address both the challenges and opportunities in human T(reg) cell therapy.


Subject(s)
Adoptive Transfer , Cell- and Tissue-Based Therapy , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory/transplantation , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Humans , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance
15.
Clin Exp Immunol ; 172(2): 169-77, 2013 May.
Article in English | MEDLINE | ID: mdl-23574314

ABSTRACT

Cell-based therapies using natural or genetically modified regulatory T cells (T(regs)) have shown significant promise as immune-based therapies. One of the main difficulties facing the further advancement of these therapies is that the fate and localization of adoptively transferred T(regs) is largely unknown. The ability to dissect the migratory pathway of these cells in a non-invasive manner is of vital importance for the further development of in-vivo cell-based immunotherapies, as this technology allows the fate of the therapeutically administered cell to be imaged in real time. In this review we will provide an overview of the current clinical imaging techniques used to track T cells and T(regs) in vivo, including magnetic resonance imaging (MRI) and positron emission tomography (PET)/single photon emission computed tomography (SPECT). In addition, we will discuss how the finding of these studies can be used, in the context of transplantation, to define the most appropriate T(reg) subset required for cellular therapy.


Subject(s)
Adoptive Transfer , Magnetic Resonance Imaging , Positron-Emission Tomography , T-Lymphocytes, Regulatory/physiology , T-Lymphocytes, Regulatory/transplantation , Tomography, Emission-Computed, Single-Photon , Animals , Cell Line , Diagnostic Imaging , Humans , Mice , Mice, Inbred C57BL , Transplantation
16.
J Thromb Haemost ; 11(5): 963-74, 2013 May.
Article in English | MEDLINE | ID: mdl-23516969

ABSTRACT

BACKGROUND: CD34(+) α-smooth muscle actin (SMA)(+) cells mediate intimal hyperplasia (IH) after mechanical endoluminal injury. We previously found that IH is tissue factor (TF) dependent. The precise phenotype of the CD34(+) cells mediating IH is unknown and the mechanisms of TF are also unknown. OBJECTIVE: To define the phenotype of cells mediating IH and compare the effects of inhibiting TF on different subsets of CD34(+) cells. METHODS: Endoluminal injury was induced in C57BL/6 and two strains of mice expressing a human tissue factor pathway inhibitor (hTFPI) fusion protein on different subsets of CD34(+) cells. Confocal microscopy, immunocytofluorescence and real-time PCR were used to determine phenotype. RESULTS: Neointimal cells in C57BL/6 mice were defined as a subset of fibrocytes (CD34(+) CD45(+) collagen-1(+) ) expressing SMA, CD31, TIE-2, CXCR4 and CXCL12. Similar cells circulated post-injury and were also found in mice expressing hTFPI on CD34(+) CD31(+) cells, though in these mice, hTFPI inhibited CD31(+) fibrocyte hyperplasia, so no IH developed. Mice with hTFPI on all CD34(+) α-SMA(+) cells repaired arteries back to a pre-injured state. No CD31(+) fibrocytes were found in these mice unless an anti-hTFPI antibody was administered. Similar findings in protease activated receptor (PAR)-1-deficient mice suggested hTFPI prevented thrombin signaling through PAR-1. In vitro, thrombin increased the number of CD31(+) fibrocytes. CONCLUSIONS: Inhibition of TF on CD31(+) fibrocytes inhibits IH whereas inhibition on all CD34(+) α-SMA(+) cells (or PAR-1 deficiency) inhibits the appearance of CD31(+) fibrocytes and promotes repair. These data enhance our understanding of IH and suggest novel ways to promote regenerative repair.


Subject(s)
Fibroblasts/pathology , Hyperplasia/pathology , Thromboplastin/physiology , Tunica Intima/pathology , Animals , Fibroblasts/immunology , Immunophenotyping , Mice , Real-Time Polymerase Chain Reaction
17.
Clin Exp Immunol ; 170(3): 300-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23121671

ABSTRACT

Helicobacter pylori is one of the most common infections in the world. Despite inciting inflammation, immunological clearance of the pathogen is often incomplete. CD4(+) CD25(hi) forkhead box protein 3 (FoxP3(+)) regulatory T cells (T(regs)) are potent suppressors of different types of immune responses and have been implicated in limiting inflammatory responses to H. pylori. Investigating the influence of H. pylori on T(reg) function and proliferation, we found that H. pylori-stimulated dendritic cells (DCs) induced proliferation in T(regs) and impaired their suppressive capability. This effect was mediated by interleukin (IL)-1ß produced by H. pylori-stimulated DCs. These data correlated with in-vivo observations in which H. pylori(+) gastric mucosa contained more T(regs) in active cell division than uninfected stomachs. Inciting local proliferation of T(regs) and inhibiting their suppressive function may represent a mechanism for the chronic gastritis and carcinogenesis attributable to H. pylori.


Subject(s)
Dendritic Cells/immunology , Helicobacter pylori/immunology , Interleukin-1beta/biosynthesis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Humans , Interleukin-6/biosynthesis , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis
18.
Am J Transplant ; 12(3): 640-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22151236

ABSTRACT

To investigate the role of donor-specific indirect pathway T cells in renal transplant tolerance, we analyzed responses in peripheral blood of 45 patients using the trans-vivo delayed-type hypersensitivity assay. Subjects were enrolled into five groups-identical twin, clinically tolerant (TOL), steroid monotherapy (MONO), standard immunosuppression (SI) and chronic rejection (CR)-based on transplant type, posttransplant immunosuppression and graft function. The indirect pathway was active in all groups except twins but distinct intergroup differences were evident, corresponding to clinical status. The antidonor indirect pathway T effector response increased across patient groups (TOL < MONO < SI < CR; p < 0.0001) whereas antidonor indirect pathway T regulatory response decreased (TOL > MONO = SI > CR; p < 0.005). This pattern differed from that seen in circulating naïve B-cell numbers and in a cross-platform biomarker analysis, where patients on monotherapy were not ranked closest to TOL patients, but rather were indistinguishable from chronically rejecting patients. Cross-sectional analysis of the indirect pathway revealed a spectrum in T-regulatory:T-effector balance, ranging from TOL patients having predominantly regulatory responses to CR patients having predominantly effector responses. Therefore, the indirect pathway measurements reflect a distinct aspect of tolerance from the recently reported elevation of circulating naïve B cells, which was apparent only in recipients off immunosuppression.


Subject(s)
B-Lymphocytes/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , Kidney Transplantation/immunology , T-Lymphocytes/immunology , Tissue Donors , Humans , Immunosuppression Therapy , Prognosis , Signal Transduction
19.
Am J Transplant ; 11(8): 1610-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21797973

ABSTRACT

CD4(+) CD25(+) regulatory T cells (T(reg) cells) are an attractive adoptive cell therapy in mediating transplantation tolerance. T-cell receptor (TcR) activation is critical for T(reg) function, suggesting that the TcR avidity of T(reg) cells used in therapy may affect the therapeutic outcome. To address this, we compared the regulatory capacity of T(reg) lines expressing TcRs derived from two TcR transgenic mice shown to have the same specificity but different functional avidities. T(reg) lines generated from CD4(+)CD25(+) T cells from C57BL/6 mice were transduced with one of either of these TcRs. The antigen specificity of the transduced T(reg) lines was confirmed in vitro. T(reg) lines expressing the TcR with higher functional avidity showed stronger suppressive capacity in a linked suppression model in vitro. Furthermore, the same T(reg) lines demonstrated a stronger proliferation in vivo following antigen exposure. Pretreatment of recipient BL/6 mice with these T(reg) cells, together with anti-CD8 antibody and Rapamycin therapies, prolonged survival of BALB/c skins, as compared with mice that received T(reg) lines with lower TcR avidity. Taken together, these data suggest that the TcR functional avidity may be important for T(reg) function. It highlights the fact that strategies to select T(reg) with higher functional avidity might be beneficial for immunotherapy in transplantation.


Subject(s)
T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , DNA Primers , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
20.
Am J Transplant ; 11(8): 1734-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21749646

ABSTRACT

Successful expansion of functional CD4(+) CD25(+) regulatory T cells (T(reg)) ex vivo under good manufacturing practice conditions has made T(reg) -cell therapy in clinical transplant tolerance induction a feasible possibility. In animals, T(reg) cells home to both transplanted tissues and local lymph nodes and are optimally suppressive if active at both sites. Therefore, they have the opportunity to suppress both naïve and memory CD4(+) CD25(-) T cells (Tresp). Clinical transplantation commonly involves depleting therapy at induction (e.g. anti-CD25), which favors homeostatic expansion of memory T cells. Animal models suggest that T(reg) cells are less suppressive on memory, compared with naïve Tresp that mediate allograft rejection. As a result, in the context of human T(reg) -cell therapy, it is important to define the effectiveness of T(reg) cells in regulating naïve and memory Tresp. Therefore, we compared suppression of peripheral blood naïve and memory Tresp by fresh and ex vivo expanded T(reg) cells using proliferation, cytokine production and activation marker expression (CD154) as readouts. With all readouts, naïve human Tresp were more suppressible by approximately 30% than their memory counterparts. This suggests that T(reg) cells may be more efficacious if administered before or at the time of transplantation and that depleting therapy should be avoided in clinical trials of T(reg) cells.


Subject(s)
CD4 Antigens/immunology , Immunologic Memory , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocytes, Regulatory/immunology , Cell Separation , Cells, Cultured , Flow Cytometry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...