Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 11: 224, 2020.
Article in English | MEDLINE | ID: mdl-32265897

ABSTRACT

To evaluate the expression of immune checkpoint genes, their concordance with expression of IFNγ, and to identify potential novel ICP related genes (ICPRG) in colorectal cancer (CRC), the biological connectivity of six well documented ("classical") ICPs (CTLA4, PD1, PDL1, Tim3, IDO1, and LAG3) with IFNγ and its co-expressed genes was examined by NGS in 79 CRC/healthy colon tissue pairs. Identification of novel IFNγ- induced molecules with potential ICP activity was also sought. In our study, the six classical ICPs were statistically upregulated and correlated with IFNγ, CD8A, CD8B, CD4, and 180 additional immunologically related genes in IFNγ positive (FPKM > 1) tumors. By ICP co-expression analysis, we also identified three IFNγ-induced genes [(IFNγ-inducible lysosomal thiol reductase (IFI30), guanylate binding protein1 (GBP1), and guanylate binding protein 4 (GBP4)] as potential novel ICPRGs. These three genes were upregulated in tumor compared to normal tissues in IFNγ positive tumors, co-expressed with CD8A and had relatively high abundance (average FPKM = 362, 51, and 25, respectively), compared to the abundance of the 5 well-defined ICPs (Tim3, LAG3, PDL1, CTLA4, PD1; average FPKM = 10, 9, 6, 6, and 2, respectively), although IDO1 is expressed at comparably high levels (FPKM = 39). We extended our evaluation by querying the TCGA database which revealed the commonality of IFNγ dependent expression of the three potential ICPRGs in 638 CRCs, 103 skin cutaneous melanomas (SKCM), 1105 breast cancers (BC), 184 esophageal cancers (ESC), 416 stomach cancers (STC), and 501 lung squamous carcinomas (LUSC). In terms of prognosis, based on Pathology Atlas data, correlation of GBP1 and GBP4, but not IFI30, with 5-year survival rate was favorable in CRC, BC, SKCM, and STC. Thus, further studies defining the role of IFI30, GBP1, and GBP4 in CRC are warranted.


Subject(s)
Breast Neoplasms/genetics , Colon/physiology , Colorectal Neoplasms/genetics , Interferon-gamma/metabolism , Melanoma/genetics , Skin Neoplasms/genetics , Stomach Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , High-Throughput Nucleotide Sequencing , Humans , Immune Checkpoint Proteins/genetics , Male , Melanoma/immunology , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Prognosis , Skin Neoplasms/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Survival Analysis , Melanoma, Cutaneous Malignant
2.
Vaccine ; 33(32): 3865-72, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26122582

ABSTRACT

Sublethal γ irradiation eliminates CD8+ T cell mediated memory responses. In this work, we explored how these memory responses could be rescued in the aftermath of such exposure. We utilized two models of CD8+ T cell mediated immunity: a mouse model of Listeria monocytogenes (LM) infection in which CD8+ T cells specific for LM expressed antigens (Listeriolysin O, LLO) can be tracked, and a murine skin graft model in which CD8+ T cells mediate rejection across a MHC class I (D(d)) disparity. In the LM immunized mice, LL0 specific CD8+ T memory cells were lost on irradiation, preserved with rapid revaccination with an attenuated strain 1-3 days post-irradiation (PI), and these mice survived a subsequent wild type LM challenge. A genetic "signature of rescue" identified a group of immune-associated mRNA maintained or upregulated following irradiation and rescue. A number of these factors, including IL-36γ, dectin-2 (Clec4n), and mir101c are upregulated rapidly after exposure of mice to sublethal γ radiation alone and are sustained by early, but not later rescue. Such factors will be evaluated as potential therapeutics to replace individual vaccines for global rescue of CD8+ T memory cell responses following sublethal γ irradiation. The skin allograft model mirrored that of the LM model in that the accelerated D(d) skin allograft rejection response was lost in mice exposed to sublethal γ radiation, but infusion of allogeneic D(d) expressing bone marrow cells 1-4 days PI preserved the CD8+ T memory mediated accelerated rejection response, further suggesting that innate immune responses may not always be essential to rescue of CD8+ memory T cells following γ irradiation.


Subject(s)
Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Gamma Rays , Immunologic Memory , Radiation Exposure , Allografts/immunology , Animals , Bacterial Vaccines/administration & dosage , Female , Graft Rejection , Listeria monocytogenes/immunology , Mice, Inbred BALB C
3.
PLoS One ; 7(6): e39092, 2012.
Article in English | MEDLINE | ID: mdl-22723935

ABSTRACT

Sublethal total body γ irradiation (TBI) of mammals causes generalized immunosuppression, in part by induction of lymphocyte apoptosis. Here, we provide evidence that a part of this immune suppression may be attributable to dysfunction of immune regulation. We investigated the effects of sublethal TBI on T cell memory responses to gain insight into the potential for loss of vaccine immunity following such exposure. We show that in mice primed to an MHC class I alloantigen, the accelerated graft rejection T memory response is specifically lost several weeks following TBI, whereas identically treated naïve mice at the same time point had completely recovered normal rejection kinetics. Depletion in vivo with anti-CD4 or anti-CD25 showed that the mechanism involved cells consistent with a regulatory T cell (T reg) phenotype. The loss of the T memory response following TBI was associated with a relative increase of CD4+CD25+ Foxp3+ expressing T regs, as compared to the CD8+ T effector cells requisite for skin graft rejection. The radiation-induced T memory suppression was shown to be antigen-specific in that a third party ipsilateral graft rejected with normal kinetics. Remarkably, following the eventual rejection of the first MHC class I disparate skin graft, the suppressive environment was maintained, with markedly prolonged survival of a second identical allograft. These findings have potential importance as regards the immunologic status of T memory responses in victims of ionizing radiation exposure and apoptosis-inducing therapies.


Subject(s)
Gamma Rays/adverse effects , Immunosuppression Therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Cycle/radiation effects , Epitopes/immunology , Forkhead Transcription Factors/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/genetics , Graft Survival/immunology , Histocompatibility Antigens Class I/immunology , Immunologic Memory , Lymphocyte Depletion , Mice , Mice, Transgenic , Skin Transplantation/immunology , Transcription, Genetic , Transplantation, Homologous/immunology , Whole-Body Irradiation
4.
Curr Protoc Immunol ; Chapter 4: 4.4.1-4.4.13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19235769

ABSTRACT

Skin allograft rejection is a test of the competence of T lymphocytes to mediate in vivo tissue destruction, which in turn reflects their role in critical functions such as anti-viral and tumor immunity. The tail-skin graft procedure described in this unit is useful predominantly because of the ease of preparation and resistance to ischemic (nonspecific) necrosis. Additionally, it is not necessary to sacrifice the donor mouse. However, rejection of tail-skin grafts should not be used to test for genetic homogeneity in breeding experiments or to detect minor histocompatibility (minor-H) antigens because tail skin is less sensitive than trunk skin in detecting such differences.


Subject(s)
Graft Rejection/immunology , Skin Transplantation/methods , Transplantation Immunology , Animals , Female , Mice , Minor Histocompatibility Antigens/immunology , Skin Transplantation/immunology , Skin Transplantation/instrumentation , T-Lymphocytes/immunology , Tail , Transplantation, Homologous
5.
Blood ; 101(11): 4520-8, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12586613

ABSTRACT

We constructed a chimeric molecule, composed of the T-cell receptor (TCR)-zeta chain fused to the extracellular domains of a prototypical allogeneic major histocompatibility complex (MHC) class I molecule, Dd, to assess whether such a construct could affect Dd allospecific responses in vitro and in vivo. To generate cytotoxic T lymphocytes (CTLs) expressing the construct, Dd-zeta was targeted to lymphocyte populations in transgenic mice by placing its expression under control of the CD2 promoter. In response to ligation of Dd, lymphocytes from transgenic mice expressing high levels of Dd-zeta are activated to proliferate and kill cells binding to Dd, despite the near total loss of CD8+ T cells in these mice. Thus, the Dd-zeta cytolytic cell was found not to be a conventional CD8+ CTL, but rather an unusual T lineage cell (CD3-CD5+Thy1.1+) that lacked alphabeta or gammadelta TCRs, as well as CD4 and CD8 coreceptors, but expressed surface markers strikingly similar to memory CTLs, including CD44, Ly-6C, and CD122. These cells originate in the thymus and potently veto responses to Dd in vitro. Lacking TCRs, these veto cells are unlikely to mediate graft-versus-host disease (GVHD) and thus may be useful as a cellular therapy for therapeutic deletion of alloreactive T cells in the settings of graft rejection and GVHD.


Subject(s)
Histocompatibility Antigens Class I/immunology , Immunologic Memory , Receptors, Antigen, T-Cell/deficiency , T-Lymphocytes, Cytotoxic/immunology , Animals , Cytotoxicity, Immunologic , Humans , Immunophenotyping , Mice , Mice, Transgenic , Recombinant Fusion Proteins , Signal Transduction , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Tumor Cells, Cultured
6.
Eur J Immunol ; 32(7): 2055-67, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12115627

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease caused by T cell-dependent antibody-mediated reduction of acetylcholine receptors (AChR) at the neuromuscular junction. Immunization of animals with Torpedo californica AChR (TAChR) results in an experimental model of MG. We used the variable regions of alpha and beta T cell receptor (TCR) genes recognizing an immunodominant peptide containing amino acids 146-162 from the alpha subunit of TAChR presented in the context of I-A(b) to generate TCR-transgenic mice. We found that the transgenic TCR was strongly positively selected and that transgenic T cells proliferated robustly to the immunodominant peptide and TAChR. Unexpectedly, there was a variable paucity of B cells in the blood and spleen from transgenic mice, which averaged about 16% of peripheral blood lymphocytes, compared to 55% in wild-type B6 mice. Unselected transgenic mice immunized with TAChR exhibited weak anti-TAChR antibody responses. However, transgenic mice selected to have relatively higher B cell numbers produced anti-TAChR titers equal to B6 mice and a predominance of Th1-induced antibody isotypes were observed in certain experiments. The incidence and severity of clinical disease was variable following immunizations. These mice should be useful for studying the pathogenesis and treatment of MG.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Cholinergic/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Division , Gene Expression , Lymphocyte Count , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/genetics , Torpedo , Vesicular stomatitis Indiana virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...