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.
Immunology ; 144(1): 91-106, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24990517

ABSTRACT

Therapies that promote tolerance in solid organ transplantation will improve patient outcomes by eliminating the need for long-term immunosuppression. To investigate mechanisms of rapamycin-induced tolerance, C3H/HeJ mice were heterotopically transplanted with MHC-mismatched hearts from BALB/cJ mice and were monitored for rejection after a short course of rapamycin treatment. Mice that had received rapamycin developed tolerance with indefinite graft survival, whereas untreated mice all rejected their grafts within 9 days. In vitro, splenic mononuclear cells from tolerant mice maintained primary CD4(+) and CD8(+) immune responses to donor antigens consistent with a mechanism that involves active suppression of immune responses. Furthermore, infection with lymphocytic choriomeningitis virus strain WE led to loss of tolerance suggesting that tolerance could be overcome by infection. Rapamycin-induced, donor-specific tolerance was associated with an expansion of regulatory T (Treg) cells in both the spleen and allograft and elevated plasma levels of fibrinogen-like protein 2 (FGL2). Depletion of Treg cells with anti-CD25 (PC61) and treatment with anti-FGL2 antibody both prevented tolerance induction. Tolerant allografts were populated with Treg cells that co-expressed FGL2 and FoxP3, whereas rejecting allografts and syngeneic grafts were nearly devoid of dual-staining cells. We examined the utility of an immunoregulatory gene panel to discriminate between tolerance and rejection. We observed that Treg-associated genes (foxp3, lag3, tgf-ß and fgl2) had increased expression and pro-inflammatory genes (ifn-γ and gzmb) had decreased expression in tolerant compared with rejecting allografts. Taken together, these data strongly suggest that Treg cells expressing FGL2 mediate rapamycin-induced tolerance. Furthermore, a gene biomarker panel that includes fgl2 can distinguish between rejecting and tolerant grafts.


Subject(s)
Fibrinogen/immunology , Graft Rejection/prevention & control , Heart Transplantation , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/drug effects , Allografts , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Fibrinogen/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Granzymes/genetics , Granzymes/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Depletion/methods , Mice , Mice, Inbred BALB C , Mice, Knockout , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transplantation Tolerance/genetics , Lymphocyte Activation Gene 3 Protein
2.
PLoS One ; 8(10): e72309, 2013.
Article in English | MEDLINE | ID: mdl-24146739

ABSTRACT

Mounting effective innate and adaptive immune responses are critical for viral clearance and the generation of long lasting immunity. It is known that production of inhibitory factors may result in the inability of the host to clear viruses, resulting in chronic viral persistence. Fibrinogen-like protein 2 (FGL2) has been identified as a novel effector molecule of CD4(+)CD25(+) Foxp3(+) regulatory T (Treg) cells that inhibits immune activity by binding to FCγRIIB expressed primarily on antigen presenting cells (APC). In this study, we show that infection of mice with Lymphocytic Choriomeningitis Virus WE (LCMV WE) leads to increased plasma levels of FGL2, which were detected as early as 2 days post-infection (pi) and persisted until day 50 pi. Mice deficient in FGL2 (fgl2(-/-)) had increased viral titers of LCMV WE in the liver early p.i but cleared the virus by day 12 similar to wild type mice. Dendritic cells (DC) isolated from the spleens of LCMV WE infected fgl2(-/-) had increased expression of the DC maturation markers CD80 and MHC Class II compared to wild type (fgl2(+/+)). Frequencies of CD8(+) and CD4(+) T cells producing IFNγ in response to ex vivo peptide re-stimulation isolated from the spleen and lymph nodes were also increased in LCMV WE infected fgl2(-/-) mice. Increased frequencies of CD8(+) T cells specific for LCMV tetramers GP33 and NP396 were detected within the liver of fgl2(-/-) mice. Plasma from fgl2(-/-) mice contained higher titers of total and neutralizing anti-LCMV antibody. Enhanced anti-viral immunity in fgl2(-/-) mice was associated with increased levels of serum alanine transaminase (ALT), hepatic necrosis and inflammation following LCMV WE infection. These data demonstrate that targeting FGL2 leads to early increased viral replication but enhanced anti-viral adaptive T & B cell responses. Targeting FGL2 may enhance the efficacy of current anti-viral therapies for hepatotropic viruses.


Subject(s)
Adaptive Immunity , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Fibrinogen/immunology , Hepatitis/genetics , Lymphocytic choriomeningitis virus/immunology , Alanine Transaminase/blood , Alanine Transaminase/genetics , Animals , Antibodies, Neutralizing/blood , B-Lymphocytes/immunology , B-Lymphocytes/virology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Female , Fibrinogen/genetics , Gene Deletion , Gene Expression Regulation , Hepatitis/immunology , Hepatitis/pathology , Hepatitis/virology , Host-Pathogen Interactions , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Mice , Mice, Knockout , Signal Transduction , Spleen/immunology , Spleen/virology , Viral Load , Virus Replication
3.
Liver Transpl ; 18(7): 761-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22523007

ABSTRACT

Liver transplantation is now recognized as the most effective therapy for patients with end-stage acute and chronic liver failure. Despite outstanding short-term graft and patient survival, liver transplantation continues to face several major challenges, including poor long-term graft survival due to chronic rejection and major side effects of long-term immunosuppressive therapy (which is required for the prevention of rejection). The ability to produce a state of tolerance after transplantation would potentially obviate long-term immunosuppression. Self-tolerance and immune homeostasis involve both central and peripheral immunoregulatory mechanisms. To date, studies have shown that many subsets of regulatory T cells (Tregs) control immune responses to foreign and alloantigens. The identification of Tregs that are positive for CD4, CD25, and the transcription factor forkhead box (Foxp3) has resulted in major advances in our understanding of the immunology of rejection and the development of transplant tolerance. In this article, we focus on the importance of Tregs in tolerance induction in experimental models of liver transplantation. Furthermore, we discuss the therapeutic potential of Tregs for the promotion of tolerance in transplant patients and highlight recent clinical trials of Treg-based therapies.


Subject(s)
Liver Transplantation/methods , T-Lymphocytes, Regulatory/cytology , Animals , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/cytology , Epigenesis, Genetic , Forkhead Transcription Factors/metabolism , Graft Rejection , Humans , Immunosuppressive Agents/therapeutic use , Interleukin-2 Receptor alpha Subunit/biosynthesis , Mice , Models, Biological , Thymus Gland/metabolism , Transplantation Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL
...