Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Am J Transplant ; 10(3): 464-71, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121738

ABSTRACT

The 10th Banff Conference on Allograft Pathology was held in Banff, Canada from August 9 to 14, 2009. A total of 263 transplant clinicians, pathologists, surgeons, immunologists and researchers discussed several aspects of solid organ transplants with a special focus on antibody mediated graft injury. The willingness of the Banff process to adapt continuously in response to new research and improve potential weaknesses, led to the implementation of six working groups on the following areas: isolated v-lesion, fibrosis scoring, glomerular lesions, molecular pathology, polyomavirus nephropathy and quality assurance. Banff working groups will conduct multicenter trials to evaluate the clinical relevance, practical feasibility and reproducibility of potential changes to the Banff classification. There were also sessions on quality improvement in biopsy reading and utilization of virtual microscopy for maintaining competence in transplant biopsy interpretation. In addition, compelling molecular research data led to the discussion of incorporation of omics-technologies and discovery of new tissue markers with the goal of combining histopathology and molecular parameters within the Banff working classification in the near future.


Subject(s)
Antibodies/chemistry , Organ Transplantation/methods , Biopsy , Canada , Complement C4b/metabolism , Fibrosis/pathology , Humans , Kidney Diseases/diagnosis , Kidney Diseases/pathology , Kidney Diseases/virology , Kidney Transplantation , Multicenter Studies as Topic , Peptide Fragments/metabolism , Phenotype , Polyomavirus Infections/diagnosis , Quality Control
2.
Am J Transplant ; 7(11): 2605-14, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17868071

ABSTRACT

The role of non-complement-activating alloantibodies in humoral graft rejection is unclear. We hypothesized that the non-complement-activating alloantibodies synergistically activate complement in combination with complement-activating antibodies. B10.A hearts were transplanted into immunoglobulin knock out (Ig-KO) mice reconstituted with monoclonal antibodies to MHC class I antigens. In allografts of unreconstituted Ig-KO recipients, no C4d was detected. Similarly, reconstitution with IgG1 or low dose IgG2b alloantibodies did not induce C4d deposition. However, mice administered with a low dose of IgG2b combined with IgG1 had heavy linear deposits of C4d on vascular endothelium. C4d deposits correlated with decreased graft survival. To replicate this synergy in vitro, mononuclear cells from B10.A mice were incubated with antibodies to MHC class I antigens followed by incubation in normal mouse serum. Flow cytometry revealed that both IgG2a and IgG2b synergized with IgG1 to deposit C4d. This synergy was significantly decreased in mouse serum deficient in mannose binding lectin (MBL) and in serum deficient in C1q. Reconstitution of MBL-A/C knock out (MBL-KO) serum with C1q-knock out (C1q-KO) serum reestablished the synergistic activity. This suggests a novel role for non-complement-activating alloantibodies and MBL in humoral rejection.


Subject(s)
Complement C4b/immunology , Complement System Proteins/immunology , Heart Transplantation/immunology , Isoantibodies/blood , Peptide Fragments/immunology , Animals , Complement Activation/immunology , Complement C4b/analysis , Flow Cytometry , Heart Transplantation/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Peptide Fragments/analysis , Rabbits , Transplantation, Homologous/immunology
3.
Am J Transplant ; 6(5 Pt 1): 923-32, 2006 May.
Article in English | MEDLINE | ID: mdl-16611328

ABSTRACT

Antibody-mediated rejection of human cardiac transplants is correlated with C4d deposits and macrophage infiltrates in capillaries of endomyocardial biopsies. We produced an antibody to rat C4d to study C4d deposition and clearance in Lewis rats that were sensitized with a blood transfusion from DA rats 7, 14 or 21 days before cardiac transplantation. Cyclosporin A (CsA) immunosuppression was initiated after transplantation at a dose that inhibited graft rejection, antibody production and C4d deposition in unsensitized recipients. Blood transfusion elicited high levels of circulating IgG alloantibodies, predominantly of the complement-activating IgG2b subclass, that peaked 14 days after transplantation. At this time, macrophages accumulated in capillaries, and C4d deposits were diffuse and intense on arteries, capillaries and veins. Grafts that survived 90 days in sensitized recipients still had deposits of C4d that were associated with increased interstitial fibrosis and vasculopathy in arteries. Clearance of C4d was determined by retransplanting DA cardiac allografts from Lewis recipients back to DA recipients. C4d deposits were decreased to minimal levels within 5 days after retransplantation. Thus, C4d deposition is not limited to the capillaries, but extends throughout the arterial tree, and despite formation of a covalent bond, C4d is cleared within days.


Subject(s)
Complement C4b/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Isoantibodies/blood , Peptide Fragments/immunology , Animals , Complement C4b/metabolism , Graft Survival/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Peptide Fragments/metabolism , Rats , Rats, Inbred Lew , Rats, Inbred Strains
4.
Transplant Proc ; 37(1): 32-4, 2005.
Article in English | MEDLINE | ID: mdl-15808537

ABSTRACT

Antibodies can mediate injury of organ transplants by several mechanisms, including complement activation and interaction with Fc receptors on cells. We tested the hypothesis that antibodies could also cause up-regulation of complement receptors on cells to increase the responses to complement activation by interaction with split products of C3. In our experimental model, B10.A (H-2(a)) cardiac transplants survive significantly longer in C57BL/6 (H-2(b)) immunoglobulin knockout recipients (IgKO) than in their wild-type counterparts. Passive transfer of specific antibodies to donor MHC class I to IgKO recipients of cardiac allografts at the time coinciding with a vigorous cellular infiltration reconstituted acute rejection. We tested the effects of alloantibodies on CR1/2 expression by alloantigen-stimulated T cells. Both CD4(+)/CR1/2(+) and CD8(+)/CR1/2(+) populations of T cells were expanded in C57BL/6 splenocytes stimulated by B10.A alloantigen in 7-day MLR after coculture with endothelial cells sensitized with IgG1 and IgG2b mAb specific to MHC. Endothelial cells sensitized with antibodies also caused an expansion of CD8(+) T cells expressing CR1/2 in lymph node lymphocytes harvested from a C57BL/6 recipient of a B10.A cardiac allograft. These data suggest that antibodies can augment the cellular rejection process through expanding the population of T cells interacting with complement split products.


Subject(s)
Heart Transplantation/immunology , Isoantigens/pharmacology , Receptors, Complement 3b/genetics , Receptors, Complement 3d/genetics , T-Lymphocytes/immunology , Animals , Cell Line , Endothelium, Vascular , Flow Cytometry , Lymph Nodes/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/drug effects
5.
Transplant Proc ; 37(1): 96-7, 2005.
Article in English | MEDLINE | ID: mdl-15808559

ABSTRACT

Previous pregnancy is a known risk factor for alloantibody production and graft rejection in clinical transplantation. However, in previous rat models, immune responses to RT1.A antigens induced by allogeneic pregnancy resulted in prolonged survival of subsequent allografts. This study was designed to investigate the effects of a previous pregnancy on alloantibody response, complement activation, and allograft survival in a highly immunogenic rat strain combination. C6-sufficient and -deficient female PVG.1U (RT1.A(u)B(u)) rats were mated with allogeneic PVG.R8 (RT1.A(a)B(u)) males or control isogeneic PVG.1U (RT1.A(u)B(u)) males. Three weeks after parturition, experimental and control females received cardiac allografts from female PVG.R8 donors. A low dose of cyclosporine (CsA, 5 mg/kg on alternate days) was used for immunosuppression after transplantation. Allogeneic, but not control isogeneic, pregnancy elicited a weak, transient IgG alloantibody response that declined before transplantation. Experimental female recipients produced a rapid, vigorous IgM and IgG alloantibody response to the transplant despite CsA treatment. C6-sufficient recipients rejected their transplants at an accelerated rate (5 days, n = 6) compared with control animals (7 days, n = 5). In contrast, allografts to C6-deficient recipients functioned until sacrifice at 90 days in both the experimental group (n = 7) and control group (n = 4). Most experimental C6-deficient recipients continued to produce strong IgG alloantibodies for 90 days. Complement activation resulting from the alloantibody response was evidenced by the diffuse deposition of C3d on the vascular endothelium of the grafts. In summary, previous pregnancy leads to memory alloantibody responses that accelerate allograft rejection even with immunosuppression. Membrane attack complex is required for accelerated rejection induced by previous pregnancy.


Subject(s)
Complement C6/immunology , Heart Transplantation/immunology , Pregnancy, Animal/immunology , Animals , Antibody Formation , Complement C6/deficiency , Complement C6/genetics , Complement Membrane Attack Complex/immunology , Female , Histocompatibility Testing , Immunization , Isoantibodies/immunology , Models, Animal , Pregnancy , Rats , Rats, Inbred Strains , Transplantation, Homologous/immunology
6.
Circulation ; 104(19): 2369-75, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11696480

ABSTRACT

BACKGROUND: Inducible nitric oxide synthase (iNOS, or NOS2) reduces the severity of accelerated graft arteriosclerosis (AGA) in transplanted organs, although the precise mechanism is unclear. METHODS AND RESULTS: We transplanted wild-type murine hearts into either wild-type or NOS2-null recipient mice; we then measured cardiac allograft survival and analyzed tissue sections by immunohistochemistry. We have confirmed that NOS2 increases cardiac allograft survival. We now show that there is less inflammation of cardiac allografts in wild-type hosts than in NOS2-null hosts. Furthermore, staining for von Willebrand factor reveals that the presence of NOS2 is correlated with the presence of Weibel-Palade bodies inside endothelial cells, whereas the absence of NOS2 is correlated with the release of Weibel-Palade bodies. CONCLUSIONS: Weibel-Palade bodies contain mediators that promote thrombosis and inflammation. Therefore, nitric oxide (NO) may stabilize the vessel wall and prevent endothelial activation in part by inhibiting the release of the contents of Weibel-Palade bodies. Prevention of Weibel-Palade body release might be a mechanism by which NO protects the vessel wall from inflammatory disorders such as atherosclerosis or graft arteriosclerosis.


Subject(s)
Graft Rejection/pathology , Heart Transplantation , Nitric Oxide Synthase/metabolism , Transplantation, Homologous/pathology , Weibel-Palade Bodies/pathology , Animals , Disease Progression , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Fluorescent Antibody Technique , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Heart Transplantation/immunology , Immunohistochemistry , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Transplantation, Homologous/immunology , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/ultrastructure , von Willebrand Factor/biosynthesis
7.
Transplantation ; 71(8): 1179-83, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11374423

ABSTRACT

BACKGROUND: Although treatment of LEW rats with Rapamycin (RPM) prolongs the survival of LBNF1 cardiac allografts to ca. 50 days, it fails to prevent late activation of macrophage/monocyte-associated chemokines. METHODS: A 7-day course with RPM or CsA was introduced at 1-week posttransplant in RPM-pretreated hosts. At day 35, intragraft mRNA expression of IL-2, IFN-gamma, TGF-beta, IL-12 (p40), MCP-1, and RANTES was evaluated. RESULTS: RPM as a sequential treatment markedly inhibited mRNA levels coding for IL-2/IFN-gamma, and MCP-1. However, RPM monotherapy failed to prevent the expression of IL-12 (p40) and RANTES. Adjunctive treatment with CsA markedly depressed IL-12 (p40) and RANTES, and to a lesser extent MCP-1 mRNA, as compared with RPM-treated groups. Significant increase of TGF-beta mRNA expression was revealed after sequential RPM and adjunctive CsA treatment. CONCLUSIONS: A combination of RPM and CsA is more effective in restraining mRNA expression than RPM alone; however, either therapy is associated with TGF-beta hyperexpression.


Subject(s)
Chemokines/genetics , Cyclosporine/therapeutic use , Cytokines/genetics , Gene Expression Regulation/immunology , Heart Transplantation/immunology , Macrophages/immunology , Monocytes/immunology , Sirolimus/therapeutic use , Animals , Gene Expression Regulation/drug effects , Immunosuppressive Agents/therapeutic use , Macrophages/drug effects , Monocytes/drug effects , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Transplantation, Isogeneic
8.
Transplantation ; 71(6): 727-36, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11330533

ABSTRACT

BACKGROUND: Alloantibody is an intrinsic component of the immune response to organ transplants. Although alloantibodies have been correlated with decreased graft survival, the mechanisms of alloantibody-mediated injury remain largely undefined in vivo. In the present study, we have established a model of alloantibody-mediated graft injury using B10.A (H-2a) hearts transplanted to wild type (WT) or immunoglobulin knock out (IgKO) C57BL-Igh-6 (H-2b) mice. METHODS: Alloantibodies were measured in the circulation and graft by flow cytometry and in immunofluorescence staining, respectively. Intragraft cytokine mRNA expression was evaluated using a competitive template reverse transcriptase polymerase chain reaction (RT-PCR) technique. P-selectin and von Willebrand factor expression were localized by immunoperoxidase staining. The capacity of alloantibodies to restore acute cardiac allograft rejection was tested by passive transfer of monoclonal antibodies (mAbs) against donor major histocompatibility complex (MHC) class I antigens to IgKO recipients. RESULTS: B10.A cardiac allografts are rejected acutely by WT C57BL/6 recipients, but over 50% of the cardiac allografts survived more than 50 days after transplantation in IgKO mice. Competitive template RT-PCR on the cardiac transplants demonstrated similar levels of IL-1-alpha, IL-12 (p40), TNF-alpha, IL-2, IFN-gamma, IL-4, and IL-10 mRNA in WT and IgKO recipients 8-10 days after transplantation, indicating that macrophage- and T-cell-dependent immune responses were intact in IgKO recipients. The rejection of B10.A hearts in WT recipients was characterized by interstitial and perivascular cellular infiltration; IgG, IgM, and complement (C3) deposition; vascular cell injury and intravascular platelet aggregation; and release of von Willebrand factor and P-selectin. In IgKO recipients the lower degree of vascular injury in the absence of alloantibody responses was reflected by the lack of release of von Willebrand factor and P-selectin, which remained confined to cytoplasmic storage granules of endothelial cells and platelets. Acute rejection of cardiac allografts was restored to IgKO recipients by passive transfer of proinflammatory IgG2b mAbs against donor MHC; recipients injected with isotype-matched control mAbs did not reject. In contrast, passive transfer of IgG1 mAbs against donor MHC failed to restore acute rejection of cardiac allografts to IgKO recipients. Passive transfer of IgG2b, but not IgG1 mAbs was associated with endothelial cell activation and plate. let aggregation together with the release of preformed von Willebrand factor and P-selectin from storage granules. CONCLUSIONS: Acute rejection of cardiac allografts can be reconstituted in IgKO recipients by passive transfer of IgG2b, but not IgG1 antibody. This model allows the mechanism of alloantibody-mediate graft injury to be dissected in vivo.


Subject(s)
Heart Transplantation/immunology , Isoantibodies/immunology , Mice, Knockout/immunology , Acute Disease , Animals , Antibody Formation , Endothelium, Vascular/cytology , Endothelium, Vascular/injuries , Graft Rejection/immunology , Graft Survival/immunology , Immunization, Passive , Immunoglobulins/deficiency , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , P-Selectin/metabolism , T-Lymphocytes/immunology , von Willebrand Factor/metabolism
10.
Am J Pathol ; 155(4): 1293-302, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10514411

ABSTRACT

The terminal components of complement C5b-C9 can cause significant injury to cardiac allografts. Using C6-deficient rats, we have found that the rejection of major histocompatibility (MHC) class I-incompatible PVG.R8 (RT1.A(a)B(u)) cardiac allografts by PVG.1U (RT1.A(u)B(u)) recipients is particularly dependent on C6. This model was selected to determine whether tissue injury results from C6 produced by macrophages, which are a conspicuous component of infiltrates in rejecting transplants. We demonstrated that high levels of C6 mRNA are expressed in isolated populations of macrophages. The relevance of macrophage-produced C6 to cardiac allograft injury was investigated by transplanting hearts from PVG. R8 (C6-) donors to PVG.1U (C6-) rats which had been reconstituted with bone marrow from PVG.1U (C6+) rats as the sole source of C6. Hearts grafted to hosts after C6 reconstitution by bone marrow transplantation underwent rejection characterized by deposition of IgG and complement on the vascular endothelium together with extensive intravascular aggregates of P-selectin-positive platelets. At the time of acute rejection, the cardiac allografts contained extensive perivascular and interstitial macrophage infiltrates. RT-PCR and in situ hybridization demonstrated high levels of C6 mRNA in the macrophage-laden transplants. C6 protein levels were also increased in the circulation during rejection. To determine the relative contribution to cardiac allograft rejection of the low levels of circulating C6 produced systemically by macrophages, C6 containing serum was passively transferred to PVG.1U (C6-) recipients of PVG.R8 (C6-) hearts. This reconstituted the C6 levels to about 3 to 6% of normal values, but failed to induce allograft rejection. In control PVG.1U (C6-) recipients that were reconstituted with bone marrow from PVG.1U (C6-) donors, C6 levels remained undetectable and PVG.R8 cardiac allografts were not rejected. These results indicate that C6 produced by macrophages can cause significant tissue damage.


Subject(s)
Complement C6/biosynthesis , Complement C6/immunology , Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Macrophages/metabolism , Animals , Animals, Congenic , Bone Marrow Transplantation , Complement C3/metabolism , Complement C3d/metabolism , Complement C6/genetics , Enzyme-Linked Immunosorbent Assay , Graft Rejection/blood , Histocompatibility Testing , Immunophenotyping , In Situ Hybridization , Isoantibodies/blood , Isoantibodies/metabolism , Macrophages/cytology , Macrophages/immunology , Male , Myocardium/pathology , P-Selectin/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction
14.
Arch Immunol Ther Exp (Warsz) ; 31(6): 809-17, 1983.
Article in English | MEDLINE | ID: mdl-6378133

ABSTRACT

The effect of a combination of specific and non-specific immunosuppression on survival of heart and skin allografts in parental strain rats differing across the major histocompatibility locus was investigated. Pretreatment of the recipient with donor specific antigen and anti-donor alloantiserum prolonged the survival of heart grafts but not skin grafts. The use of antilymphocyte serum prolonged skin graft survival but not heart graft survival. However, when ATS was combined with donor specific immunosuppression, both heart and skin graft survival were prolonged dramatically. It is suggested that these treatment modalities affect differentially various aspects of the immune responses.


Subject(s)
Antilymphocyte Serum/immunology , Heart Transplantation , Immunization, Passive , Immunosuppression Therapy , Skin Transplantation , T-Lymphocytes/immunology , Animals , Cytotoxicity, Immunologic , Graft Survival , Immune Tolerance , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL