Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Cell Immunol ; 347: 104023, 2020 01.
Article in English | MEDLINE | ID: mdl-31836133

ABSTRACT

Autologous C-kit+ cells robustly prolong cardiac allografts. As C-kit+ cells can transdifferentiate to hematopoietic cells as well as non-hematopoietic cells, we aimed to clarify the class(es) of C-kit-derived cell(s) required for cardiac allograft prolongation. Autologous C-kit+ cells were administered post-cardiac transplantation and allografts were evaluated for C-kit+ inoculum-derived cells. Results suggested that alloimmunity was a major signal for trafficking of C-kit-derived cells to the allograft and demonstrated that C-kit+ inoculum-derived cells expressed CD11b early after transfer. Allograft survival studies with CD11b-DTR C-kit+ cells demonstrated a requirement for C-kit+-derived CD11b+ cells. Co-therapy studies demonstrated near complete abrogation of acute rejection with concomitant CTLA4-Ig therapy and no loss of prolongation in combination with Cyclosporine A. These results strongly implicate a C-kit-derived myeloid population as critical for allograft preservation and demonstrate the potential therapeutic application of autologous C-kit+ progenitor cells as calcineurin inhibitor-sparing agents and possibly as co-therapeutics for durable graft survival.


Subject(s)
CD11b Antigen/metabolism , Graft Survival/immunology , Heart Transplantation/methods , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Transplantation , Abatacept/pharmacology , Allografts , Animals , Calcineurin Inhibitors , Cardiomyopathies/mortality , Cardiomyopathies/surgery , Cyclosporine/pharmacology , Female , Graft Rejection/immunology , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/immunology , Stem Cells/physiology , Transplantation, Homologous
2.
Am J Transplant ; 17(7): 1742-1753, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28066981

ABSTRACT

Several approaches successfully achieve allograft tolerance in preclinical models but are challenging to translate into clinical practice. Many clinically relevant factors can attenuate allograft tolerance induction, including intrinsic genetic resistance, peritransplant infection, inflammation, and preexisting antidonor immunity. The prevailing view for immune memory as a tolerance barrier is that the host harbors memory cells that spontaneously cross-react to donor MHC antigens. Such preexisting "heterologous" memory cells have direct reactivity to donor cells and resist most tolerance regimens. In this study, we developed a model system to determine if an alternative form of immune memory could also block tolerance. We posited that host memory T cells could potentially respond to donor-derived non-MHC antigens, such as latent viral antigens or autoantigens, to which the host is immune. Results show that immunity to a model nonself antigen, ovalbumin (OVA), can dramatically disrupt tolerance despite undetectable initial reactivity to donor MHC antigens. Importantly, this blockade of tolerance was CD8+ T cell-dependent and required linked antigen presentation of alloantigens with the test OVA antigen. As such, this pathway represents an unapparent, or "incognito," form of immunity that is sufficient to prevent tolerance and that can be an unforeseen additional immune barrier to clinical transplant tolerance.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Immunologic Memory/immunology , Islets of Langerhans Transplantation/methods , Transplantation Tolerance/immunology , Animals , Female , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Transplantation, Homologous
3.
Am J Transplant ; 16(11): 3105-3114, 2016 11.
Article in English | MEDLINE | ID: mdl-27516154

ABSTRACT

From June 11-15, 2016 the American Transplant Congress, the joint meeting of the American Society of Transplantation and the American Society of Transplant Surgeons, held its annual meeting in Boston, MA. The meeting, attended by 5200 registrants, included pre-meeting conferences, focused topic sessions, and hundreds of high-quality presentations from the transplant field. This meeting report highlights key findings from specific basic science, translational, and clinical research presentations deemed to have notable impact in thematic areas. In particular, there were a number of transformative studies indicating important advances in the understanding of alloimmunity, chronic rejection, tolerance, and organ-specific outcomes. Many of these results are discussed in the context of the published literature to showcase rapid advances in the transplant field.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/immunology , Immune Tolerance/immunology , Organ Transplantation/trends , Tissue and Organ Procurement/trends , Congresses as Topic , Humans , United States
4.
Am J Transplant ; 16(11): 3121-3130, 2016 11.
Article in English | MEDLINE | ID: mdl-27163757

ABSTRACT

Natural killer (NK) cells are key components of the innate immune system. In murine cardiac transplant models, donor-specific antibodies (DSA), in concert with NK cells, are sufficient to inflict chronic allograft vasculopathy independently of T and B cells. In this study, we aimed to determine the effector mechanism(s) required by NK cells to trigger chronic allograft vasculopathy during antibody-mediated rejection. Specifically, we tested the relative contribution of the proinflammatory cytokine interferon gamma (IFN-γ) versus the contact-dependent cytotoxic mediators of perforin and the CD95/CD95L (Fas/Fas ligand [FasL]) pathway for triggering these lesions. C3H/HeJ cardiac allografts were transplanted into immune-deficient C57BL/6 rag-/- γc-/- recipients, who also received monoclonal anti-major histocompatibility complex (MHC) class I DSA. The combination of DSA and wild-type NK cell transfer triggered aggressive chronic allograft vasculopathy. However, transfer of IFN-γ-deficient NK cells or host IFN-γ neutralization led to amelioration of these lesions. Use of either perforin-deficient NK cells or CD95 (Fas)-deficient donors alone did not alter development of vasculopathy, but simultaneous disruption of NK cell-derived perforin and allograft Fas expression resulted in prevention of these abnormalities. Therefore, both NK cell IFN-γ production and contact-dependent cytotoxic activity are rate-limiting effector pathways that contribute to this form of antibody-induced chronic allograft vasculopathy.


Subject(s)
Antibodies, Monoclonal/immunology , Graft Rejection/immunology , Heart Transplantation , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cytokines/metabolism , Fas Ligand Protein/immunology , Female , Histocompatibility Antigens Class I/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Perforin/immunology , Transplantation, Homologous , fas Receptor/immunology
5.
Transplant Proc ; 46(6): 2007-9, 2014.
Article in English | MEDLINE | ID: mdl-25131095

ABSTRACT

BACKGROUND: Despite ongoing advances in the clinical islet transplant field, progressive decline in graft function continues to reduce the long-term success of islet transplantation for restoring euglycemia in type 1 diabetic recipients. To preserve graft function and avoid the use of chronic immunosuppressive drug therapy, a key goal is to induce donor-specific immune tolerance to islet transplants. Preclinical rodent studies of islet transplantation largely utilize models of diabetes either induced experimentally with beta cell toxins or spontaneously occurring in strains genetically prone to autoimmune diabetes. In this study, we sought to determine if chronic, severe hyperglycemia itself, independent of both beta cell toxins and host autoimmunity, influenced acute allograft rejection and/or the capacity to induce allograft tolerance. METHOD: To this end, we studied the response to islet allografts in severely diabetic, non-autoimmune C57Bl/6 Ins2(akita) recipients. RESULTS: Results indicate that diabetic Ins2(akita) mice display higher levels of blood glucose, show more rapid acute islet allograft rejection, and are resistant to allograft prolongation induced with anti-CD154 therapy relative to wild-type littermates rendered diabetic with streptozotocin. As such, results suggest that severe hyperglycemia may be an independent risk factor impacting the capacity to induce tolerance to islet allografts. Thus, Ins2(akita) mice represent a stringent model for evaluating anti-rejection strategies in the setting of severe metabolic demand on islet transplants.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD40 Ligand/immunology , Diabetes Mellitus, Experimental/surgery , Immunologic Factors/therapeutic use , Islets of Langerhans Transplantation , Transplantation Tolerance , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Graft Survival/immunology , Male , Mice , Mice, Inbred C57BL , Transplantation, Homologous
7.
Am J Transplant ; 11(1): 34-44, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21114653

ABSTRACT

Autologous CD117(+) progenitor cells (PC) have been successfully utilized in myocardial infarction and ischemic injury, potentially through the replacement/repair of damaged vascular endothelium. To date, such cells have not been used to enhance solid organ transplant outcome. In this study, we determined whether autologous bone marrow-derived CD117(+) PC could benefit cardiac allograft survival, possibly by replacing donor vascular cells. Autologous, positively selected CD117(+) PC were administered posttransplantation and allografts were assessed for acute rejection. Although significant generation of recipient vascular cell chimerism was not observed, transferred PC disseminated both to the allograft and to peripheral lymphoid tissues and facilitated a significant, dose-dependent prolongation of allograft survival. While CD117(+) PC dramatically inhibited alloreactive T cell proliferation in vitro, this property did not differ from nonprotective CD117(-) bone marrow populations. In vivo, CD117(+) PC did not significantly inhibit T cell alloreactivity or increase peripheral regulatory T cell numbers. Thus, rather than inhibiting adaptive immunity to the allograft, CD117(+) PC may play a cytoprotective role in prolonging graft survival. Importantly, autologous CD117(+) PC appear to be distinct from bone marrow-derived mesenchymal stem cells (MSC) previously used to prolong allograft survival. As such, autologous CD117(+) PC represent a novel cellular therapy for promoting allograft survival.


Subject(s)
Heart Transplantation/immunology , Proto-Oncogene Proteins c-kit/immunology , Stem Cells/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation/immunology , Graft Survival/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , T-Lymphocytes/immunology , Transplantation, Homologous
9.
Transplant Proc ; 40(2): 462-3, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374102

ABSTRACT

Islet allografts are destroyed rapidly in spontaneously diabetic nonobese diabetic (NOD) mice. However, whether this process is more similar to conventional allograft immunity, islet-specific autoimmune pathogenesis, or both remains controversial. In particular, we sought to determine whether C57BI/6 donor islet major histocompatibility complex (MHC) class I or class II expression was required for islet allograft destruction in autoimmune prone NOD mice versus non-autoimmune-prone BALB/c mice. Results show that islet allografts deficient in both MHC I and II are uniformly accepted in BALB/c mice. In sharp contrast, such MHC-deficient allografts were destroyed acutely in spontaneously diabetic NOD mice. Such donor MHC-independent rejection implicates "indirect" (host MHC-restricted) immunity as a pathway responsible for islet injury. To determine whether host NOD B lymphocytes could contribute to indirect graft recognition, wild-type and MHC I/II-deficient allografts were grafted into B-lymphocyte-deficient (microMT) NOD mice. Whereas wild-type NOD mice could reject MHC-I/II-deficient islet allografts, such grafts were all accepted in B-lymphocyte-deficient NOD mice. Taken together, these results indicate that NOD mice are capable of vigorous donor MHC-independent islet allograft rejection not found in non-autoimmune-prone recipients. Importantly, B lymphocytes may play a key role as antigen-presenting cells in this exuberant host 'indirect' response found in NOD mice.


Subject(s)
B-Lymphocytes/immunology , Islets of Langerhans Transplantation/pathology , Major Histocompatibility Complex , Animals , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Transplantation, Homologous
10.
Transplant Proc ; 40(2): 557-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374128

ABSTRACT

Natural killer (NK) cells have long been appreciated for their rapid, proinflammatory contribution to host defense. However, more recent studies show an unexpected regulatory role for host major histocompatibility complex (MHC) class I-dependent immunity and NK cells in promoting tolerance induction to islet allografts. It is unclear whether the potential tolerance induction to islet xenografts follows similar requirements to those found in allograft tolerance. In this study, we determined whether induced islet xenograft prolongation also showed a reliance on MHC class I-dependent immune pathways. In particular, we tested whether NK1.1+ cells and/or CD8 T cells were required for the long-term islet xenograft survival in a rat-to-mouse transplant model. Short-term host treatment with combined anti-CD154 plus anti-LFA-1 (CD11a) resulted in prolonged, but not indefinite, survival of WF rat islet xenografts in C57BI/6 mouse recipients. In stark contrast with similar islet allograft studies, adjunct treatment with anti-NK1.1 therapy combined wither anti-CD154/anti-LFA-1 treatment led to long-term (>100 days) survival of the majority of islet xenografts. In parallel studies, we determined whether CD8 T cells also contributed a barrier to xenograft survival. Similar to results found in anti-NK1.1-treated animals, CD8-deficient (knockout) recipients also demonstrated augmented xenograft prolongation after combined anti-CD154/anti-LFA-1 therapy. Taken together, NK1.1+ cells (NK/NKT cells) and CD8 T cells constitute differing MHC class I-dependent immune pathways forming a significant barrier to xenograft prolongation.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Innate , Islets of Langerhans Transplantation/immunology , Killer Cells, Natural/immunology , Transplantation, Heterologous/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Primates , Swine
11.
Am J Transplant ; 7(8): 1927-33, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17617855

ABSTRACT

Though CD8(+) T lymphocytes are important cellular mediators of islet allograft rejection, their molecular mechanism of rejection remains unidentified. Surprisingly, while it is generally assumed that CD8(+) T cells require classic cytotoxic mechanisms to kill grafts in vivo, neither perforin nor FasL (CD95L) are required for acute islet allograft rejection. Thus, it is unclear whether such contact-dependent cytotoxic pathways play an essential role in islet rejection. Moreover, both perforin and CD95L have been implicated in playing roles in peripheral tolerance, further obscuring the role of these effector pathways in rejection. Therefore, we determined whether perforin and/or FasL (CD95L) were required by donor MHC-restricted ('direct') CD8(+) T cells to reject islet allografts in vivo. Islet allograft rejection by primed, alloreactive CD8(+) T cells was examined independently of other lymphocyte subpopulations via adoptive transfer studies. Individual disruption of T-cell-derived perforin or allograft Fas expression had limited impact on graft rejection. However, simultaneous disruption of both pathways prevented allograft rejection in most recipients despite the chronic persistence of transferred T cells at the graft site. Thus, while there are clearly multiple cellular pathways of allograft rejection, perforin and FasL comprise alternate and necessary routes of acute CD8(+) T-cell-mediated islet allograft rejection.


Subject(s)
Fas Ligand Protein/metabolism , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Membrane Glycoproteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Acute Disease , Adoptive Transfer/methods , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Female , Genes, MHC Class I/physiology , Graft Rejection/metabolism , Graft Rejection/pathology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Perforin , Prognosis , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Transplantation, Homologous
12.
Diabetologia ; 50(8): 1649-59, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17593347

ABSTRACT

AIMS/HYPOTHESIS: Transplantation of islets is a viable option for the treatment of diabetes. A significant proportion of islets is lost during isolation, storage and after transplantation as a result of apoptosis. cAMP response element binding protein (CREB) is an important cell survival factor. The aim of the present study was to determine whether preservation of CREB function is needed for survival of human islets. MATERIALS AND METHODS: To determine the effects of downregulation of CREB activity on beta cell apoptosis in a transplantation setting, adenoviral vectors were used to express two dominant negative mutant forms of CREB in human islets isolated from cadaveric donors. Markers of apoptosis were determined in these transduced islets under basal conditions and following treatment with growth factor. RESULTS: Expression of CREB mutants in human islets resulted in significant (p < 0.001) activation of caspase-9, a key regulatory enzyme in the mitochondrial pathway of apoptosis, when compared with islets transduced with adenoviral beta galactosidase. Immunocytochemical analysis showed the activation of caspase-9 to be predominantly in beta cells. Other definitive markers of apoptosis such as parallel activation of caspase-3, accumulation of cleaved poly-(ADP-ribose) polymerase and nuclear condensation were also observed. Furthermore, the anti-apoptotic action of growth factors exendin-4 and betacellulin in human islets exposed to cytokines was partially lost when CREB function was impaired. CONCLUSIONS/INTERPRETATION: Our findings suggest that impairment of CREB-mediated transcription could lead to loss of islets by apoptosis with potential implications in islet transplantation as well as in the mechanism of beta cell loss leading to diabetes.


Subject(s)
Apoptosis/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Islets of Langerhans/drug effects , Mutation , Adenoviridae/genetics , Apoptosis/genetics , Apoptosis/physiology , Betacellulin , Cadaver , Caspase 9/metabolism , Caspases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/physiology , Cytokines/pharmacology , Exenatide , Genes, Dominant , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Islets of Langerhans/metabolism , Peptides/pharmacology , Transfection , Venoms/pharmacology
13.
J Clin Endocrinol Metab ; 91(5): 1855-61, 2006 May.
Article in English | MEDLINE | ID: mdl-16478822

ABSTRACT

CONTEXT: Type 1A diabetes is characterized by a long prodromal phase during which autoantibodies to islet antigens are present. Nevertheless, we lack data on the pancreatic pathology of subjects who are positive for islet autoantibodies (to islet autoantigens GAD65, insulin, and ICA512). OBJECTIVE: In this manuscript, we describe a novel strategy in obtaining pancreata and pancreatic lymph nodes from islet autoantibody-positive organ donors that involves careful coordination among the laboratory and the organ donor provider organization. DESIGN: We developed a rapid screening protocol for islet autoantibodies measurement of organ donors to allow identification of positive subjects before organ harvesting. In this way we were able to obtain pancreata and pancreatic lymph nodes from subjects with and without islet autoimmunity. SETTING: The organ donors used in this study were obtained from the general community. SUBJECTS: The population studied consisted of 112 organ donors (age range 1 month to 86 yr, mean age 39 yr). MAIN OUTCOME MEASURE: The main outcome measure of this study consisted of evaluating the pancreatic histology and identify T cells autoreactive for islet antigens in the pancreatic lymph nodes. RESULTS: To date we have identified three positive subjects and obtained the pancreas for histological evaluation from one of the autoantibody-positive donors who expressed ICA512 autoantibodies. Although this subject did not exhibit insulitis, lymphocytes derived from pancreatic lymph nodes reacted to the islet antigen phogrin. CONCLUSION: In summary, these results indicate that it is possible to screen organ donors in real time for antiislet antibodies, characterize pancreatic histology, and obtain viable T cells for immunological studies.


Subject(s)
Autoantibodies/analysis , Islets of Langerhans/immunology , Tissue Donors , Adolescent , Adult , Aged , Chromogranins/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibrosis , Glucagon/analysis , Glucagon/metabolism , Humans , Immunohistochemistry , Insulin/analysis , Insulin/metabolism , Islets of Langerhans/pathology , Keratins/metabolism , Leukocyte Common Antigens/analysis , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Middle Aged , Pancreas/metabolism , T-Lymphocytes/immunology
14.
Am J Transplant ; 6(1): 27-36, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16433753

ABSTRACT

Leukocyte function associated antigen-1 (LFA-1) was one of the earliest of cell-surface molecules identified by monoclonal antibodies generated against leukocyte immunogens. This integrin heterodimer is perhaps best known as a classic adhesion molecule facilitating the interaction between T cells and antigen-presenting cells. However, varied studies indicate that LFA-1 has multi-faceted roles in the immune response including adhesion, activation and trafficking of leukocyte populations. While there has been long-standing interest in LFA-1 as a therapeutic target for regulating immunity, anti-LFA-1 therapy is still not a first-line indication for any clinical condition. Antagonism of LFA-1 with monoclonal antibodies, either alone or in combination with other agents, can result in regulatory tolerance in vivo. Furthermore, new generation humanized anti-LFA-1 monoclonal antibodies (Efalizumab) show at least modest promise for continued application in clinical trials. Thus, anti-LFA-1 forms a potential, but still largely unexploited, immunotherapy which may find its greatest application as an agent which augments other therapies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD11a Antigen/drug effects , Graft Rejection/prevention & control , Lymphocyte Function-Associated Antigen-1/drug effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , CD11a Antigen/immunology , Graft Rejection/immunology , Humans , Leukocyte Rolling/drug effects , Leukocyte Rolling/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Organ Transplantation
15.
Transplant Proc ; 36(4): 1171-2, 2004 May.
Article in English | MEDLINE | ID: mdl-15194405

ABSTRACT

The role of B7 family members CD80 and CD86 in providing costimulatory signals to T cells is well established. Interestingly, previous studies show that host CD80/CD86 expression is required for cardiac allograft rejection. However, the role for host costimulation by CD80/CD86 molecules for the rejection of neovascularized islet allografts and xenografts is unknown. The purpose of this study was to determine whether islet allografts and/or rat islet xenografts required host CD80/CD86 molecules for acute rejection. Streptozotocin-induced diabetic C57Bl/6 (B6, H-2(b)) or B6 CD80/CD86 double-deficient mice were grafted with allogeneic BALB/c (H-2(d)) islet allografts or with WF (RT1(u)) islet xenografts. Nondiabetic B6 mice were grafted with BALB/c heterotopic cardiac allografts. Consistent with previous reports, BALB/c islet allografts were acutely rejected in wild-type B6 mice could survive long-term (>100 days) in B6 CD80/CD86-deficient animals. In stark contrast, both islet allografts and WF rat islet xenografts demonstrated acute rejection in both control B6 and in B6 CD80/CD86 deficient hosts. In conclusion, varied studies imply that the inherent pathways for rejecting primarily vascularized versus cellular allografts or xenografts may be distinct. The present study illustrates this concept by showing a marked difference in the role of host-derived CD80/CD86 costimulatory molecules for cardiac allograft versus islet allograft/xenograft rejection in vivo. Although such costimulation is rate limiting for cardiac allograft rejection, these same molecules are not necessary for acute rejection of either islet allografts or xenografts.


Subject(s)
Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Major Histocompatibility Complex/immunology , Animals , CD40 Ligand/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Animal , Transplantation, Homologous/immunology
16.
Transplant Proc ; 36(4): 1173-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15194406

ABSTRACT

The purpose of this study was to determine the role for CD8 T cells versus generalized MHC class I-restricted antigen presentation in islet allograft rejection and tolerance. Diabetic C57BI/6 (B6, H-2(b)) controls, C57BI/6 CD8-deficient (CD8 KO), or MHC class I-deficient C57BI/6 (beta 2m KO) recipients were grafted with allogeneic BALB/c (H-2(d)) islets. Islet allografts were acutely rejected in untreated B6, CD8 KO, and in beta 2m KO mice, indicating that neither CD8 T cells nor host MHC class I is required for allograft rejection. We then determined the efficacy of costimulation blockade in these same strains. Costimulation blockade with anti-CD154 therapy facilitated long-term islet allograft survival in both B6 and in CD8 KO recipients. However, anti-CD154 treated beta 2m KO recipients were completely refractory to anti-CD154 therapy; all treated animals acutely rejected islet allografts with or without therapy. Also, anti-NK1.1 treatment of wild-type B6 mice abrogated graft prolongation following anti-CD154 therapy. Taken together, results show a dramatic distinction between two forms of MHC class I-restricted pathways in allograft prolongation. Although anti-CD154-induced allograft survival was CD8 T-cell independent, an intact host MHC class I-restricted (beta 2m-dependent) pathway is nevertheless necessary for allograft survival. This pathway required NK1.1+ cells, implicating NK and/or NKT cells in promoting allograft prolongation in vivo.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Histocompatibility Antigens Class I/immunology , Islets of Langerhans Transplantation/immunology , Major Histocompatibility Complex , Transplantation, Homologous/immunology , Animals , Graft Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
17.
Crit Rev Immunol ; 21(1-3): 87-101, 2001.
Article in English | MEDLINE | ID: mdl-11642616

ABSTRACT

Two signals are required for optimal T-cell activation: the engagement of the antigen-specific receptor and the provision of a second non-antigen-specific inductive signal, or costimulator (CoS). Regarding allograft immunity, two primary pathways of donor antigen presentation can fulfill this two-signal requirement, resulting in cellular immunity to a transplant: (1) "direct" (donor MHC-restricted) presentation in which the antigen-presenting cells (APCs) resident within the transplant directly activate host T lymphocytes and (2) "indirect" (host MHC-restricted) presentation in which host-derived APCs acquire donor antigens that are then presented to host T lymphocytes. It appears that endocrine allografts, such as pancreatic islets and thyroid, are highly dependent on donor-derived APCs, or "passenger leukocytes," to trigger acute graft rejection. Tissue pretreatment aimed at selectively eliminating APCs within endocrine tissues can result in indefinite allograft survival in immune-competent recipients. Although such results implicate the "direct" pathway as the predominant route of host sensitization, the role of donor APCs in rejection appears to be more complex. Recently, we have found that indirect, CD4 T-cell-dependent reactivity can contribute to islet allograft rejection. However, such indirect recognition nevertheless requires donor-derived APCs as a source of antigen. Thus, whereas the donor-type APC is a critical limiting step for initiating islet allograft rejection, such cells can trigger both direct and indirect forms of immune responses that can result in graft rejection. That is, donor hematopoietic cells, rather than tissue parenchymal cells, probably play a major role in providing antigens that stimulate cellular immunity.


Subject(s)
Antigen-Presenting Cells/physiology , Endocrine Glands/transplantation , Transplantation, Homologous/immunology , Animals , Antigen Presentation , Humans , Immune Tolerance , Lymphocyte Activation , T-Lymphocytes/immunology
18.
J Immunol ; 167(9): 5457-63, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673565

ABSTRACT

Recent studies using mouse models demonstrate that CD4(+) T cells are sufficient to mediate acute cardiac allograft rejection in the absence of CD8(+) T cells and B cells. However, the mechanistic basis of CD4-mediated rejection is unclear. One potential mechanism of CD4-mediated rejection is via elaboration of proinflammatory cytokines such as IFN-gamma. To determine whether IFN-gamma is a critical cytokine in CD4-mediated acute cardiac allograft rejection, we studied whether the expression of IFN-gamma receptors on the donor heart was required for CD4-mediated rejection. To investigate this possibility, purified CD4(+) T cells were transferred into immune-deficient mice bearing heterotopic cardiac allografts from IFN-gamma receptor-deficient (GRKO) donors. While CD4(+) T cells triggered acute rejection of wild-type heart allografts, they failed to trigger rejection of GRKO heart allografts. The impairment in CD4-mediated rejection of GRKO hearts appeared to primarily involve the efferent phase of the immune response. This conclusion was based on the findings that GRKO stimulator cells provoked normal CD4 proliferation in vitro and that intentional in vivo challenge of CD4 cells with wild-type donor APC or the adoptive transfer of in vitro primed CD4 T cells failed to provoke acute rejection of GRKO allografts. In contrast, unseparated lymph node cells acutely rejected both GRKO and wild-type hearts with similar time courses, illustrating the existence of both IFN-gamma-dependent and IFN-gamma-independent mechanisms of acute allograft rejection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft Rejection , Heart Transplantation/immunology , Receptors, Interferon/physiology , Animals , CD4 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/physiology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transplantation, Homologous , Interferon gamma Receptor
19.
Transplantation ; 71(11): 1549-55, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11435963

ABSTRACT

BACKGROUND: Adhesion molecules are involved in multiple steps of the continuum of allograft rejection. We studied the effects of blockade of the interactions between alpha4 integrin and its ligands, vascular cell adhesion molecule-1 (VCAM-1) and fibronectin, on allograft survival. METHODS: Streptozotocin-induced diabetic CBA (H-2k) mice received islet transplants from BALB/c (H-2d) donors. Recipient mice were treated with antibodies against alpha4 integrin (PS/2), VCAM-1 (MK 2.7), and a peptide corresponding to the binding site of alpha4 integrin on fibronectin (connecting segment 1 peptide, CS1-peptide). Graft function was measured by daily tail vein blood glucose levels, with rejection defined as the return of hyperglycemia. Graft-bearing kidneys were removed for immunohistochemical analysis. RESULTS: Treatment with anti-alpha4 integrin antibody, anti-VCAM-1 antibody, or with CS1-peptide led to long-term survival of islet allografts. Recipients with long-surviving islet grafts did not show tolerance, in that they rejected a second donor-type islet allograft. Although both anti-alpha4 integrin antibody and CS1-peptide completely abolished cellular infiltration of the islet graft 7 days after transplantation, anti-VCAM-1-treated recipients showed a dense peri-islet infiltrate of activated, alpha4 integrin+, cytotoxic T cells. CONCLUSIONS: These data show that alpha4 integrin is critically important to allograft rejection. Anti-VCAM-1 antibody appears to prevent rejection without qualitatively affecting either T cell activation or migration to the graft. Conversely, anti-alpha4 integrin antibody and CS1-peptide may prevent islet allograft rejection by altering either T cell activation or lymphocyte trafficking. Blocking interactions between alpha4 integrin and its ligands may provide novel forms of immunosuppression.


Subject(s)
Antigens, CD/metabolism , Graft Rejection/metabolism , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Animals , Antibodies/pharmacology , Antigens, CD/immunology , Graft Survival , Immune Tolerance , Integrin alpha4 , Intercellular Signaling Peptides and Proteins , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/immunology , Lymphocyte Culture Test, Mixed , Lymphocytes/drug effects , Lymphocytes/pathology , Lymphocytes/physiology , Male , Mice , Mice, Inbred BALB C , Peptides/pharmacology , Reoperation , Time Factors , Vascular Cell Adhesion Molecule-1/immunology
20.
Article in English | MEDLINE | ID: mdl-11460981

ABSTRACT

Heart transplantation has been performed clinically for four decades, and has become the standard of care for end-stage heart disease. Our understanding of the immunobiology of transplantation has made tremendous strides, but our knowledge still lags behind the clinical use. As a result, nonspecific immunosuppression remains the standard therapy. This chapter is a review of our present knowledge of the immunobiology of allotransplantation and xenotransplantation with emphasis on antigen presentation, costimulation, and T-cell activation in the context of transplantation. The molecular events of T-cell activation, with some emphasis on the sites of action of present day immunosuppression, are reviewed. Basic aspects of immunosuppression are reviewed elsewhere in this edition. Given the paucity of allografts, xenografts are being considered as an alternative donor source. This being the case, cellular and humoral response to xenografts is considered and contrasted with our understanding of allograft immunity. Basic mechanisms of tolerance are discussed, with examples of experimental tolerance induction in small and large animals. A brief description of special considerations for the immunology in human neonate/infant recipients is mentioned. Understanding the immunobiology of transplantation is key to making decisions regarding heart transplant recipients today, in addition to developing better protocols and the induction of tolerance in the future.


Subject(s)
Heart Transplantation/immunology , Transplantation Immunology , Transplantation, Heterologous/immunology , Transplantation, Homologous/immunology , Animals , Antibodies, Monoclonal/immunology , Antigen Presentation/immunology , Graft Rejection/immunology , Histocompatibility/immunology , Humans , Immunosuppressive Agents/therapeutic use , Infant , Infant, Newborn , Killer Cells, Natural/immunology , Mice , Rats , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...