Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 384
1.
Am J Transplant ; 16(7): 2158-71, 2016 07.
Article En | MEDLINE | ID: mdl-26749226

Facial transplantation is a life-changing procedure for patients with severe composite facial defects. However, skin is the most immunogenic of all transplants, and better understanding of the immunological processes after facial transplantation is of paramount importance. Here, we describe six patients who underwent full facial transplantation at our institution, with a mean follow-up of 2.7 years. Seum, peripheral blood mononuclear cells, and skin biopsy specimens were collected prospectively, and a detailed characterization of their immune response (51 time points) was performed, defining 47 immune cell subsets, 24 serum cytokines, anti-HLA antibodies, and donor alloreactivity on each sample, producing 4269 data points. In a nonrejecting state, patients had a predominant T helper 2 cell phenotype in the blood. All patients developed at least one episode of acute cellular rejection, which was characterized by increases in interferon-γ/interleukin-17-producing cells in peripheral blood and in the allograft's skin. Serum monocyte chemotactic protein-1 level was significantly increased during rejection compared with prerejection time points. None of the patients developed de novo donor-specific antibodies, despite a fourfold expansion in T follicular helper cells at 1 year posttransplantation. In sum, facial transplantation is frequently complicated by a codominant interferon-γ/interleukin-17-mediated acute cellular rejection process. Despite that, medium-term outcomes are promising with no evidence of de novo donor-specific antibody development.


Facial Transplantation/adverse effects , Graft Rejection/diagnosis , Graft Survival/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Th1 Cells/immunology , Adult , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Humans , Kidney Function Tests , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Prognosis , Risk Factors , Transplant Recipients
2.
Am J Transplant ; 16(1): 121-36, 2016 Jan.
Article En | MEDLINE | ID: mdl-26260101

Identification of biomarkers that assess posttransplant risk is needed to improve long-term outcomes following heart transplantation. The Clinical Trials in Organ Transplantation (CTOT)-05 protocol was an observational, multicenter, cohort study of 200 heart transplant recipients followed for the first posttransplant year. The primary endpoint was a composite of death, graft loss/retransplantation, biopsy-proven acute rejection (BPAR), and cardiac allograft vasculopathy (CAV) as defined by intravascular ultrasound (IVUS). We serially measured anti-HLA- and auto-antibodies, angiogenic proteins, peripheral blood allo-reactivity, and peripheral blood gene expression patterns. We correlated assay results and clinical characteristics with the composite endpoint and its components. The composite endpoint was associated with older donor allografts (p < 0.03) and with recipient anti-HLA antibody (p < 0.04). Recipient CMV-negativity (regardless of donor status) was associated with BPAR (p < 0.001), and increases in plasma vascular endothelial growth factor-C (OR 20; 95%CI:1.9-218) combined with decreases in endothelin-1 (OR 0.14; 95%CI:0.02-0.97) associated with CAV. The remaining biomarkers showed no relationships with the study endpoints. While suboptimal endpoint definitions and lower than anticipated event rates were identified as potential study limitations, the results of this multicenter study do not yet support routine use of the selected assays as noninvasive approaches to detect BPAR and/or CAV following heart transplantation.


Biomarkers/metabolism , Coronary Artery Disease/diagnosis , Graft Rejection/diagnosis , Heart Diseases/surgery , Heart Transplantation/adverse effects , Adult , Blotting, Western , Case-Control Studies , Clinical Trials as Topic , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Endothelin-1/metabolism , Female , Gene Expression Profiling , Graft Rejection/etiology , Graft Rejection/metabolism , Humans , Male , Middle Aged , Prospective Studies , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A
4.
Am J Transplant ; 14(12): 2869-73, 2014 Dec.
Article En | MEDLINE | ID: mdl-25387427

Lineage (CD3e, CD11b, GR1, B220 and Ly-76) negative hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) infiltrate islet allografts within 24 h posttransplantation. In fact, lineage(negative) Sca-1(+) cKit(+) ("LSK") cells, a classic signature for HSCs, were also detected among these graft infiltrating cells. Lineage negative graft infiltrating cells are functionally multi-potential as determined by a standard competitive bone marrow transplant (BMT) assay. By 3 months post-BMT, both CD45.1 congenic, lineage negative HSCs/HPCs and classic "LSK" HSCs purified from islet allograft infiltrating cells, differentiate and repopulate multiple mature blood cell phenotypes in peripheral blood, lymph nodes, spleen, bone marrow and thymus of CD45.2 hosts. Interestingly, "LSK" HSCs also rapidly infiltrate syngeneic islet transplants as well as allogeneic cardiac transplants and sham surgery sites. It seems likely that an inflammatory response, not an adaptive immune response to allo-antigen, is responsible for the rapid infiltration of islet and cardiac transplants by biologically active HSCs/HPCs. The pattern of hematopoietic differentiation obtained from graft infiltrating HSCs/HPCs, cells that are recovered from inflammatory sites, as noted in the competitive BMT assay, is not precisely the same as that of intramedullary HSCs. This does not refute the obvious multi-lineage potential of graft infiltrating HSCs/HPCs.


Bone Marrow Transplantation , Bone Marrow/immunology , Cell Lineage , Heart Transplantation , Hematopoietic Stem Cells/immunology , Islets of Langerhans Transplantation , Stem Cells/immunology , Animals , Cell Differentiation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Stem Cells/cytology , Transplantation, Homologous , Transplantation, Isogeneic
5.
Am J Transplant ; 14(7): 1543-51, 2014 Jul.
Article En | MEDLINE | ID: mdl-24913821

The nonimmunologic loss of islets in the pre-, peri-, and early post-islet transplant periods is profound. To determine the potential role that transplantation of only a marginal mass of functioning beta cells may play in triggering late nonimmunologic graft loss, we studied the effect of treatment with alpha-1-antitrypsin (AAT) in the autologous cynomolgus islet transplant model. A marginal mass of autologous islets, that is islets prepared from 70% to 80% of the pancreas, was transplanted at 1600-4100 IEQ/kg into subtotal pancreatectomized, streptozotocin-treated and insulin-deficient diabetic hosts. In this marginal mass islet transplant model, islet function is insidiously lost over time and diabetes recurs in all untreated monkeys by 180 days posttransplantation. Short-term treatment with AAT, an acute phase reactant, in the peri-transplant period serves to terminate inflammation through effects upon expression of TGFß, NFκB and AKT and favorably altering expression of cell death and survival pathways, as detected by a system biology approach and histology. These effects enabled functional expansion of the islet mass in transplanted hosts such that graft function improves rather than deteriorating over time.


Diabetes Mellitus, Experimental/therapy , Graft Rejection/prevention & control , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , alpha 1-Antitrypsin/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Haplorhini , Insulin/metabolism , Transplantation, Autologous
6.
Am J Transplant ; 13(7): 1891-7, 2013 Jul.
Article En | MEDLINE | ID: mdl-23802725

Gene expression profiling of transplant recipient blood and urine can potentially be used to monitor graft function, but the multitude of protocols in use make sharing data and comparing results from different laboratories difficult. The goal of this study was to evaluate the performance of current methods of RNA isolation, reverse transcription and quantitative polymerase chain reaction (qPCR) and to test whether multiple centers using a standardized protocol can obtain the same results. Samples, reagents and detailed instructions were distributed to six participating sites that performed RNA isolation, reverse transcription and qPCR for 18S, PRF, GZB, IL8, CXCL9 and CXCL10 as instructed. All data were analyzed at a single site. All sites demonstrated proficiency in RNA isolation and qPCR analysis. Gene expression measurements for all targets and samples had correlations >0.938. The coefficient of variation of fold-changes between pairs of samples was less than 40%. All sites were able to accurately quantify a control sample of known concentration within a factor of 1.5. Collectively, we have formulated and validated detailed methods for measuring gene expression in blood and urine that can yield consistent results in multiple laboratories.


Gene Expression Profiling/standards , Gene Expression Regulation , Kidney Transplantation , RNA, Messenger/analysis , RNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Gene Expression Profiling/methods , Humans , Limit of Detection , RNA, Messenger/genetics , RNA-Directed DNA Polymerase/genetics , Sensitivity and Specificity , Transplantation, Homologous
7.
Am J Transplant ; 13(1): 36-44, 2013 Jan.
Article En | MEDLINE | ID: mdl-23016759

Leptin, an adipose-secreted hormone, links metabolism and immunity. Our aim was to determine whether leptin affects the alloimmune response. We used an allogeneic skin transplant model as a means to analyze the allograft immune response in Lep(ob/ob) and wild-type mice. Leptin deficiency results in an increased frequency of Treg and Th2 cells and a prolonged graft survival. These effects of leptin deficiency indicate the importance of leptin and obesity in modulating the allograft immune responses. Our data suggest a possible explanation for the increased susceptibility of hyperleptinemic obese patients to acute and chronic graft rejection.


Graft Survival/physiology , Leptin/physiology , Th2 Cells/immunology , Animals , Flow Cytometry , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Transplantation, Homologous
8.
Am J Transplant ; 12(5): 1296-302, 2012 May.
Article En | MEDLINE | ID: mdl-22390179

Preclinical studies in nonhuman primates (NHP) are particularly useful to evaluate the safety and efficacy of new therapeutic proteins developed for use in clinical transplantation. We hypothesized that a treatment that selectively destroys activated cytopathic donor reactive T cells while sparing resting and immunoregulatory T cells in a mouse model might also produce long-term drug-free engraftment and tolerance without the hazards of lymphopenia in the challenging nonhuman primate islet allograft model. Short-term treatment with a regimen consisting of rapamycin, and IL-2.Ig plus mutant antagonist-type IL-15.Ig cytolytic fusion proteins (triple therapy) posttransplantation results in prolonged, drug-free engraftment of cynomolgus islet allografts. Moreover slow progressive loss of islet function in some recipients was not associated with obvious pathologic evidence of rejection.


Diabetes Mellitus, Experimental/therapy , Graft Rejection/prevention & control , Graft Survival , Interleukin-15/administration & dosage , Interleukin-2/administration & dosage , Sirolimus/therapeutic use , T-Lymphocytes/immunology , Animals , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Graft Rejection/etiology , Graft Rejection/mortality , Immune Tolerance , Immunosuppressive Agents/therapeutic use , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Islets of Langerhans Transplantation/adverse effects , Macaca fascicularis , Mice , Survival Rate , T-Lymphocytes/drug effects , Tissue Donors , Transplantation, Homologous
9.
Am J Transplant ; 12(2): 330-40, 2012 Feb.
Article En | MEDLINE | ID: mdl-22053723

The presence of alloreactive memory T cells is a major barrier for induction of tolerance in primates. In theory, delaying conditioning for tolerance induction until after organ transplantation could further decrease the efficacy of the regimen, since preexisting alloreactive memory T cells might be stimulated by the transplanted organ. Here, we show that such "delayed tolerance" can be induced in nonhuman primates through the mixed chimerism approach, if specific modifications to overcome/avoid donor-specific memory T-cell responses are provided. These modifications include adequate depletion of CD8+ memory T cells and timing of donor bone marrow administration to minimize levels of proinflammatory cytokines. Using this modified approach, mixed chimerism was induced successfully in 11 of 13 recipients of previously placed renal allografts and long-term survival without immunosuppression could be achieved in at least 6 of these 11 animals.


Bone Marrow Transplantation/immunology , Graft Survival/immunology , Immunologic Memory/immunology , Kidney Transplantation/immunology , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Transplantation Tolerance/immunology , Animals , Bone Marrow Transplantation/pathology , Disease Models, Animal , Flow Cytometry , Follow-Up Studies , Kidney Transplantation/pathology , Macaca fascicularis , Male , Transplantation Conditioning/methods , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
10.
Am J Transplant ; 10(11): 2410-20, 2010 Nov.
Article En | MEDLINE | ID: mdl-20977632

We have shown that CD39 and CD73 are coexpressed on the surface of murine CD4+ Foxp3+ regulatory T cells (Treg) and generate extracellular adenosine, contributing to Treg immunosuppressive activity. We now describe that CD39, independently of CD73, is expressed by a subset of blood-derived human CD4+ CD25+ CD127lo Treg, defined by robust expression of Foxp3. A further distinct population of CD4+ CD39+ T lymphocytes can be identified, which do not express CD25 and FoxP3 and exhibit the memory effector cellular phenotype. Differential expression of CD25 and CD39 on circulating CD4+ T cells distinguishes between Treg and pathogenic cellular populations that secrete proinflammatory cytokines such as IFNγ and IL-17. These latter cell populations are increased, with a concomitant decrease in the CD4+ CD25+ CD39+ Tregs, in the peripheral blood of patients with renal allograft rejection. We conclude that the ectonucleotidase CD39 is a useful and dynamic lymphocytes surface marker that can be used to identify different peripheral blood T cell-populations to allow tracking of these in health and disease, as in renal allograft rejection.


Antigens, CD/biosynthesis , Apyrase/biosynthesis , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Pyrophosphatases/immunology , T-Lymphocytes, Regulatory/immunology , Graft Rejection/immunology , Humans , Immunologic Memory , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Kidney Failure, Chronic/immunology , Kidney Transplantation , Phenotype , Pyrophosphatases/biosynthesis , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
11.
Am J Transplant ; 9(10): 2303-11, 2009 Oct.
Article En | MEDLINE | ID: mdl-19656134

Foxp3(+) regulatory T cells (Tregs) express both ectoenzymes CD39 and CD73, which in tandem hydrolyze pericellular ATP into adenosine, an immunoinhibitory molecule that contributes to Treg suppressive function. Using Foxp3GFP knockin mice, we noted that the mouse CD4(+)CD39(+) T-cell pool contains two roughly equal size Foxp3(+) and Foxp3(-) populations. While Foxp3(+)CD39(+) cells are CD73(bright) and are the bone fide Tregs, Foxp3(-)CD39(+) cells do not have suppressive activity and are CD44(+)CD62L(-)CD25(-)CD73(dim/-), exhibiting memory cell phenotype. Functionally, CD39 expression on memory and Treg cells confers protection against ATP-induced apoptosis. Compared with Foxp3(-)CD39(-) naïve T cells, Foxp3(-)CD39(+) cells freshly isolated from non-immunized mice express at rest significantly higher levels of mRNA for T-helper lineage-specific cytokines IFN-gamma (Th1), IL-4/IL-10 (Th2), IL-17A/F (Th17), as well as pro-inflammatory cytokines, and rapidly secrete these cytokines upon stimulation. Moreover, the presence of Foxp3(-)CD39(+) cells inhibits TGF-beta induction of Foxp3 in Foxp3(-)CD39(-) cells. Furthermore, when transferred in vivo, Foxp3(-)CD39(+) cells rejected MHC-mismatched skin allografts in a much faster tempo than Foxp3(-)CD39(-) cells. Thus, besides Tregs, CD39 is also expressed on pre-existing memory T cells of Th1-, Th2- and Th17-types with heightened alloreactivity.


Antigens, CD/metabolism , Apyrase/metabolism , CD4-Positive T-Lymphocytes/metabolism , Immunologic Memory , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Knock-In Techniques , Immunophenotyping , Mice , Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
12.
Am J Transplant ; 7(7): 1722-32, 2007 Jul.
Article En | MEDLINE | ID: mdl-17511761

The outcome of T-cell-mediated responses, immunity or tolerance, critically depends on the balance of cytopathic versus regulatory T (T(reg)) cells. In the creation of stable tolerance to MHC incompatible allografts, reducing the unusually large mass of donor-reactive cytopathic T effector (T(eff)) cells via apoptosis is often required. Cyclosporine (CsA) blocks activation-induced cell death (AICD) of T(eff) cells, and is detrimental to tolerance induction by costimulation blockade, whereas Rapamycin (RPM) preserves AICD, and augments the potential of costimulation blockade to create tolerance. While differences between CsA and RPM in influencing apoptosis of activated graft-destructive T(eff) cells are apparent, their effects on graft-protective T(reg) cells remain enigmatic. Moreover, it is unclear whether tolerizing regimens foster conversion of naïve peripheral T cells into alloantigen-specific T(reg) cells for graft protection. Here we show, using reporter mice for T(reg) marker Foxp3, that RPM promotes de novo conversion of alloantigen-specific T(reg) cells, whereas CsA completely inhibits this process. Upon transfer, in vivo converted T(reg) cells potently suppress the rejection of donor but not third party skin grafts. Thus, the differential effects of RPM and CsA on T(eff) and T(reg) cells favor the use of RPM in shifting the balance of aggressive to protective type alloimmunity.


Immunosuppressive Agents/pharmacology , Isoantigens/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cyclosporine , Forkhead Transcription Factors/immunology , Genetic Markers , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sirolimus , T-Lymphocytes, Regulatory/drug effects
13.
Am J Transplant ; 7(5): 1055-61, 2007 May.
Article En | MEDLINE | ID: mdl-17286617

Heterologous immunologic memory has been considered a potent barrier to tolerance induction in primates. Induction of such tolerance for a previously transplanted organ may be more difficult, because specific memory cells can be induced and activated by a transplanted organ. In the current study, we attempted to induce tolerance to a previously transplanted kidney allograft in nonhuman primates. The conditioning regimen consisted of low dose total body irradiation, thymic irradiation, antithymocyte globulin, and anti-CD154 antibody followed by a brief course of a calcineurin inhibitor. This regimen had been shown to induce mixed chimerism and allograft tolerance when kidney transplantation (KTx) and donor bone marrow transplantation (DBMT) were simultaneously performed. However, the same regimen failed to induce mixed chimerism when delayed DBMT was performed after KTx. We found that significant levels of memory T cells remained after conditioning, despite effective depletion of naïve T cells. By adding humanized anti-CD8 monoclonal antibody (cM-T807), CD8 memory T cells were effectively depleted and these recipients successfully achieved mixed chimerism and tolerance. The current studies provide 'proof of principle' that the mixed chimerism approach can induce renal allograft tolerance, even late after organ transplantation if memory T-cell function is adequately controlled.


Antibodies, Monoclonal/pharmacology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/drug effects , Kidney Transplantation/methods , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , Antilymphocyte Serum/pharmacology , Biopsy , CD146 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Chimerism , Graft Survival/immunology , Kidney/immunology , Kidney/pathology , Kidney Transplantation/immunology , Kidney Transplantation/pathology , Macaca fascicularis , Male , Thymus Gland/radiation effects , Transplantation, Homologous , Whole-Body Irradiation
14.
Am J Transplant ; 6(4): 687-96, 2006 Apr.
Article En | MEDLINE | ID: mdl-16539625

Conventional drug therapy and several anti-CD154 mAb-based regimens were tested in the nonhuman primate (NHP) islet allograft model and found to be inadequate because islets were lost to rejection. Short-term therapy with an optimized donor-specific transfusion (DST) + rapamycin (RPM) + anti-CD154 mAb regimen enables immunosuppression drug-free islet allograft function for months following cessation of therapy in the NHP islet allograft model. After a substantial period of drug-free graft function, these allografts slowly and progressively lost function. Pathologic studies failed to identify islet allograft rejection as a destructive islet invasive lymphocytic infiltration of the allograft was not detected. To evaluate the mechanism, immunologic versus nonimmunologic, of the late islet allograft loss in hosts receiving the optimized therapeutic regimen, we performed experiments with islet autografts and studied islet function in NHPs with partial pancreatectomy. The results in both experiments utilizing autologous islet allografts and partially pancreatectomized hosts reinforce the view that the presence of a marginal islet mass leads to slowly progressive nonimmunological islet loss. Long-term clinically successful islet cell transplantation cannot be realized in the absence of parallel improvements in tolerizing regimens and in the preparation of adequate numbers of islets.


CD40 Ligand/drug effects , Diabetes Mellitus, Type 1/surgery , Graft Rejection/etiology , Graft Rejection/prevention & control , Immunosuppressive Agents/administration & dosage , Islets of Langerhans Transplantation , Animals , Antibodies/administration & dosage , Glucose Tolerance Test , Graft Rejection/pathology , Graft Survival , Macaca fascicularis , Pancreas/surgery , Pancreatectomy , Sirolimus/administration & dosage
15.
Inmunología (1987) ; 23(2): 231-238, abr. 2004. ilus
Article En | IBECS | ID: ibc-37266

La inducción de tolerancia a los aloinjertos, definida como un estado en el cual de manera selectiva el sistema inmunitario no responde frente a aloantígenos del donante en ausencia de la administración de tratamiento inmunosupresor crónico, es uno de los principales objetivos de la investigación en transplante. Múltiples evidencias indican que el balance entre linfocitos T aloagresivos y reguladores es uno de los puntos claves en el desarrollo de rechazo o bien tolerancia. Así pues, la consecución del estado de tolerancia require tanto la eliminación de un número substancial de linfocitos efectores aloreactivos, como la expansión de los mecanismos inmunoreguladores donante específicos. Las redes inmunoreguladoras activas en los receptores tolerantes se caracterizan por ser donante específicas, mediar el fenómeno de supresión ligada y depender de la vía indirecta de reconocimiento aloantigénico.Varios subtipos linfocitarios han sido implicados en los fenómenos inmunoreguladores en el transplante, pero los linfocitos T reguladores CD4+CD25+ (TReg) juegan un papel central en la inducción de tolerancia a los aloinjertos. Así, no es possible transferir la tolerancia a receptores inmunodeficientes si las células T CD4+CD25+ han sido eliminadas de entre los linfocitos tolerantes. Igualmente, la depleción de los linfocitos T CD4+CD25+ antes de la administración de tratamientos tolerogénicos puede impedir la inducción de tolerancia. Estos efectos parecen estar mediados, al menos en parte, por la capacidad de las células T CD4+CD25+ de los receptores tolerantes de incrementar su capacidad inmunosupresora de manera aloantígeno específica. Sin embargo, desconocemos todavía si las células CD4+CD25+ tienen también capacidad de mediar otros fenómenos característicos del estado de tolerancia, como por ejemplo la supresión ligada o la tolerancia infecciosa. Así mismo, no está suficientemente estudiado de qué manera las células T CD4+CD25+ interaccionan con otros tipos de linfocitos reguladores activos en la tolerancia a los transplantes (AU)


Humans , Transplantation , Graft Rejection/immunology , Immune Tolerance , CD4 Antigens/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Transplantation Immunology
17.
Transplantation ; 72(10): 1705-8, 2001 Nov 27.
Article En | MEDLINE | ID: mdl-11726838

BACKGROUND: The purpose of the present study was to investigate transcripts of perforin, granzyme B, and Fas ligand (FasL) in heart transplants undergoing rejection. METHODS: Quantitative reverse transcriptase-polymerase chain reaction was applied for mRNA detection in 29 endomyocardial biopsy specimens from 11 cardiac allograft recipients. RESULTS: The mRNA levels of granzyme B, perforin, and FasL were higher (P<0.05) in biopsy specimens with rejection than in biopsy specimens without rejection (granzyme B, 0.53 vs. 0.09; perforin, 0.34 vs. 0; FasL, 0.57 vs. 0.36). In prerejection biopsy specimens, granzyme B and FasL levels were significantly higher than in biopsy specimens without rejection. Any two of the three transcripts were increased in 100% of prerejection, in 92% of rejection, and in 36% of no rejection biopsy specimens (P<0.04). CONCLUSIONS: The assessment of intragraft levels of cytotoxic T lymphocyte effector molecule mRNA represents a valuable tool in the monitoring of cardiac allograft rejection, especially considering the predictive value for warning of impending acute rejection.


Gene Expression Regulation , Graft Rejection , Heart Transplantation/immunology , Membrane Glycoproteins/genetics , Serine Endopeptidases/genetics , T-Lymphocytes, Cytotoxic/physiology , Fas Ligand Protein , Granzymes , Humans , Perforin , Pore Forming Cytotoxic Proteins , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
20.
J Immunol ; 167(6): 3478-85, 2001 Sep 15.
Article En | MEDLINE | ID: mdl-11544341

IL-15 is a powerful T cell growth factor (TCGF) with particular importance for the maintenance of CD8(+) T cells. Because costimulation blockade does not result in universal tolerance, we hypothesized that "escape" from costimulation blockade might represent a CD8(+) and IL-15/IL-15R(+)-dependent process. For this analysis, we have used an IL-15 mutant/Fcgamma2a protein, a potentially cytolytic protein that is also a high-affinity receptor site specific antagonist for the IL-15Ralpha receptor protein, as a therapeutic agent. The IL-15-related fusion protein was used as monotherapy or in combination with CTLA4/Fc in murine islet allograft models. As monotherapies, CTLA4/Fc and an IL-15 mutant/Fcgamma2a were comparably effective in a semiallogeneic model system, and combined treatment with IL-15 mutant/Fcgamma2a plus CTLA4/Fc produced universal permanent engraftment. In a fully MHC-mismatched strain combination known to be refractory to costimulation blockade treatment, combined treatment with both fusion proteins proved to be highly effective; >70% of recipients were tolerized. The analysis revealed that the IL-15 mutant/Fc treatment confers partial protection from both CD4(+) and CD8(+) T cell graft infiltration. In rejections occurring despite CTLA4/Fc treatment, concomitant treatment with the IL-15 mutant/Fcgamma2a protein blocked a CD8(+) T cell-dominated rejection processes. This protection was linked to a blunted proliferative response of alloreactive T cells as well silencing of CTL-related gene expression events. Hence, we have demonstrated that targeting the IL-15/IL-15R pathway represents a new and potent strategy to prevent costimulation blockade-resistant CD8(+) T cell-driven rejection.


CD8-Positive T-Lymphocytes/drug effects , Graft Rejection/prevention & control , Immunoconjugates , Immunosuppressive Agents/therapeutic use , Interleukin-15/therapeutic use , Islets of Langerhans Transplantation/immunology , Lymphocyte Activation/drug effects , Recombinant Fusion Proteins/therapeutic use , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Crosses, Genetic , Diabetes Mellitus, Experimental/surgery , Drug Evaluation, Preclinical , Gene Silencing , Graft Rejection/immunology , Graft Survival/drug effects , Graft Survival/immunology , H-2 Antigens/immunology , Immune Tolerance , Immunosuppressive Agents/pharmacology , Interleukin-15/genetics , Islets of Langerhans Transplantation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Receptors, Interleukin-15 , Receptors, Interleukin-2/drug effects , Receptors, Interleukin-2/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Streptozocin , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous/immunology
...