Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Transplantation ; 88(12): 1341-8, 2009 Dec 27.
Article in English | MEDLINE | ID: mdl-20029330

ABSTRACT

BACKGROUND: Rat lung allograft rejection is mediated by collagen type V (col(V)) specific T-helper-cell 17 (Th17) cells. Adoptive transfer of these cells is sufficient to induce rejection pathology in isografts, whereas tolerance to col(V) suppresses allograft rejection. Therefore, we tested whether regulatory T cells from tolerant rats could suppress the Th17-mediated rejection in the syngeneic model of lung transplantation. METHODS: Rats were subjected to syngeneic left lung transplantation, and acute rejection was induced by adoptive transfer of lymph node cells from col(V)-immunized rats. Tolerance was induced by intravenous injection of col(V), and spleen lymphocytes were used for adoptive transfer. CD4+ T cells were depleted using magnetic beads. Lung isografts were analyzed using micro-positron emission tomography imaging and histochemistry. The transvivo delayed type hypersensitivity assay was used to analyze the Th17 response. RESULTS: Adoptive cotransfer of col(V)-specific effector cells with cells from col(V)-tolerized rats suppressed severe vasculitis and bronchiolitis with parenchymal inflammation, and the expression of interleukin (IL)-17 transcripts in mediastinal lymph nodes induced by effector cells alone. Analysis by transvivo delayed type hypersensitivity showed that the reactivity to col(V) was dependent on the presence of tumor necrosis factor-alpha and IL-17 but not interferon-gamma. Depletion of CD4+ T cells from the suppressor cell population abrogated the col(V)-specific protection. CONCLUSION: Th17-mediated acute rejection after lung transplantation is ameliorated by CD4+ col(V)-specific regulatory T cells. The mechanism for this Th17 suppression is consistent with tolerance induction to col(V). The goal of transplantation treatment, therefore, should target Th17 development and not suppression of T-cell activation by suppressing IL-2.


Subject(s)
Collagen Type V/immunology , Graft Rejection/immunology , Immunity, Cellular , Interleukin-17/immunology , Lung Transplantation/immunology , T-Lymphocyte Subsets/immunology , Acute Disease , Animals , Disease Models, Animal , Graft Rejection/diagnosis , Immunohistochemistry , Interleukin-17/biosynthesis , Positron-Emission Tomography , Rats , Rats, Inbred F344 , Rats, Inbred WKY , Transplantation, Homologous
2.
Blood ; 114(17): 3578-87, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19700665

ABSTRACT

In mice and humans, the immunologic effects of developmental exposure to noninherited maternal antigens (NIMAs) are quite variable. This heterogeneity likely reflects differences in the relative levels of NIMA-specific T regulatory (T(R)) versus T effector (T(E)) cells. We hypothesized that maintenance of NIMA-specific T(R) cells in the adult requires continuous exposure to maternal cells and antigens (eg, maternal microchimerism [MMc]). To test this idea, we used 2 sensitive quantitative polymerase chain reaction (qPCR) tests to detect MMc in different organs of NIMA(d)-exposed H2(b) mice. MMc was detected in 100% of neonates and a majority (61%) of adults; nursing by a NIMA+ mother was essential for preserving MMc into adulthood. MMc was most prevalent in heart, lungs, liver, and blood, but was rarely detected in unfractionated lymphoid tissues. However, MMc was detectable in isolated CD4+, CD11b+, and CD11c+ cell subsets of spleen, and in lineage-positive cells in heart. Suppression of delayed type hypersensitivity (DTH) and in vivo lymphoproliferation correlated with MMc levels, suggesting a link between T(R) and maternal cell engraftment. In the absence of neonatal exposure to NIMA via breastfeeding, MMc was lost, which was accompanied by sensitization to NIMA in some offspring, indicating a role of oral exposure in maintaining a favorable T(R) > T(E) balance.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chimerism , H-2 Antigens/physiology , Hypersensitivity, Delayed/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Crosses, Genetic , Female , Flow Cytometry , Hypersensitivity, Delayed/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mothers , Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism
3.
J Immunol ; 179(10): 6749-61, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982065

ABSTRACT

We hypothesize that developmental exposure to noninherited maternal Ags (NIMA) results in alloantigen-specific natural and adaptive T regulatory (T(R)) cells. We compared offspring exposed to maternal H-2(d) (NIMA(d)) with nonexposed controls. In vitro assays did not reveal any differences in T cell responses pretransplant. Adoptive transfer assays revealed lower lymphoproliferation and greater cell surface TGF-beta expression on CD4(+) T cells of NIMA(d)-exposed vs control splenocytes. NIMA(d)-exposed splenocytes exhibited bystander suppression of tetanus-specific delayed-type hypersensitivity responses, which was reversed with Abs to TGF-beta and IL-10. Allospecific T effector cells were induced in all mice upon i.v. challenge with B6D2F1 splenocytes or a DBA/2 heart transplant, but were controlled in NIMA(d)-exposed mice by T(R) cells to varying degrees. Some (40%) NIMA(d)-exposed mice accepted a DBA/2 allograft while others (60%) rejected in delayed fashion. Rejector and acceptor NIMA(d)-exposed mice had reduced T effector responses and increased Foxp3(+) T(R) cells (CD4(+)CD25(+)Foxp3(+) T(R)) in spleen and lymph nodes compared with controls. The key features distinguishing NIMA(d)-exposed acceptors from all other mice were: 1) higher frequency of IL-10- and TGF-beta-producing cells primarily in the CD4(+)CD25(+) T cell subset within lymph nodes and allografts, 2) a suppressed delayed-type hypersensitivity response to B6D2F1 Ags, and 3) allografts enriched in LAP(+), Foxp3(+), and CD4(+) T cells, with few CD8(+) T cells. We conclude that the beneficial NIMA effect is due to induction of NIMA-specific T(R) cells during ontogeny. Their persistence in the adult, and the ability of the host to mobilize them to the graft, may determine whether NIMA-specific tolerance is achieved.


Subject(s)
Graft Rejection/immunology , H-2 Antigens/immunology , Heart Transplantation/immunology , Histocompatibility, Maternal-Fetal/immunology , Isoantigens/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance , Adoptive Transfer , Animals , Bystander Effect/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Graft Rejection/metabolism , H-2 Antigens/biosynthesis , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/metabolism , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Mice , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism , Tetanus Toxoid/immunology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/immunology , Transplantation Immunology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...