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 Oncol ; 11: 662723, 2021.
Article in English | MEDLINE | ID: mdl-35223446

ABSTRACT

Myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) contribute to cancer-related inflammation and tumor progression. While several myeloid molecules have been ascribed a regulatory function in these processes, the triggering receptors expressed on myeloid cells (TREMs) have emerged as potent modulators of the innate immune response. While various TREMs amplify inflammation, others dampen it and are emerging as important players in modulating tumor progression-for instance, soluble TREM-1 (sTREM-1), which is detected during inflammation, associates with disease progression, while TREM-2 expression is associated with tumor-promoting macrophages. We hypothesized that TREM-1 and TREM-2 might be co-expressed on tumor-infiltrating myeloid cells and that elevated sTREM-1 associates with disease outcomes, thus representing a possibility for mutual modulation in cancer. Using the 4T1 breast cancer model, we found TREM-1 and TREM-2 expression on MDSC and TAM and that sTREM-1 was elevated in tumor-bearing mice in multiple models and correlated with tumor volume. While TREM-1 engagement enhanced TNF, a TREM-2 ligand was detected on MDSC and TAM, suggesting that both TREM could be functional in the tumor setting. Similarly, we detected TREM-1 and Trem2 expression in myeloid cells in the RENCA model of renal cell carcinoma (RCC). We confirmed these findings in human disease by demonstrating the expression of TREM-1 on tumor-infiltrating myeloid cells from patients with RCC and finding that sTREM-1 was increased in patients with RCC. Finally, The Cancer Genome Atlas analysis shows that TREM1 expression in tumors correlates with poor outcomes in RCC. Taken together, our data suggest that manipulation of the TREM-1/TREM-2 balance in tumors may be a novel means to modulate tumor-infiltrating myeloid cell phenotype and function.

2.
PLoS One ; 7(3): e33303, 2012.
Article in English | MEDLINE | ID: mdl-22428016

ABSTRACT

The liver is an immunologically unique organ containing tolerogenic dendritic cells (DC) that maintain an immunosuppressive microenvironment. Although systemic IL-12 administration can improve responses to tumors, the effects of IL-12-based treatments on DC, in particular hepatic DC, remain incompletely understood. In this study, we demonstrate systemic IL-12 administration induces a 2-3 fold increase in conventional, but not plasmacytoid, DC subsets in the liver. Following IL-12 administration, hepatic DC became more phenotypically and functionally mature, resembling the function of splenic DC, but differed as compared to their splenic counterparts in the production of IL-12 following co-stimulation with toll-like receptor (TLR) agonists. Hepatic DCs from IL-12 treated mice acquired enhanced T cell proliferative capabilities similar to levels observed using splenic DCs. Furthermore, IL-12 administration preferentially increased hepatic T cell activation and IFNγ expression in the RENCA mouse model of renal cell carcinoma. Collectively, the data shows systemic IL-12 administration enables hepatic DCs to overcome at least some aspects of the inherently suppressive milieu of the hepatic environment that could have important implications for the design of IL-12-based immunotherapeutic strategies targeting hepatic malignancies and infections.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/drug effects , Interleukin-12/pharmacology , Liver/immunology , Animals , Dendritic Cells/immunology , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Liver/cytology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Statistics, Nonparametric , T-Lymphocytes/immunology
3.
J Immunol ; 185(1): 174-82, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20525892

ABSTRACT

Our previous study showed that TNFR2 is preferentially expressed by CD4(+)FoxP3(+) regulatory T cells (Tregs), and expression of this receptor identified maximally suppressive Tregs. TNFR2 is also expressed by a small fraction of CD4(+)FoxP3(-) conventional T cells (Tconvs) in normal mice, and its expression is upregulated by T cell activation. This raises questions about the role of TNFR2 signaling in the function of Tconv cells. In this study, by using FoxP3/gfp knock-in mice, we showed that TNFR2 signaling did not induce FoxP3(-) CD4 cells to become suppressive. Ki-67, a marker of proliferation, was concomitantly expressed with TNFR2 by CD4 cells, independent of forkhead box P3 expression, in normal mice and Lewis lung carcinoma-bearing mice. TNFR2 is associated with greater suppressive functions when expressed by Tregs and is associated with greater resistance to suppression when expressed by Tconv cells. In mice bearing 4T1 breast tumor or Lewis lung carcinoma, intratumoral Tconv cells expressing elevated levels of TNFR2 acquired the capacity to resist suppression by lymph node-derived Tregs. However, they remained susceptible to inhibition by more suppressive tumor-infiltrating Tregs, which expressed higher levels of TNFR2. Our data indicate that TNFR2 also costimulates Tconv cells. However, intratumoral Tregs expressing more TNFR2 are able to overcome the greater resistance to suppression of intratumoral Tconv cells, resulting in a dominant immunosuppressive tumor environment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Forkhead Transcription Factors , Immunity, Innate , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/prevention & control , Cell Line, Tumor , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Coculture Techniques , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Gene Knock-In Techniques , Immunity, Innate/genetics , Lymphocyte Activation/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type II/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
4.
Eur J Immunol ; 40(4): 1099-106, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127680

ABSTRACT

Previously, we found that co-expression of CD25 and TNFR2 identified the most suppressive subset of mouse Treg. In this study, we report that human peripheral blood (PB) FOXP3(+) cells present in CD25(high), CD25(low) and even CD25(-) subsets of CD4(+) cells expressed high levels of TNFR2. Consequently, TNFR2-expressing CD4(+)CD25(+) Treg included all of the FOXP3(+) cells present in the CD4(+)CD25(high) subset as well as a substantial proportion of the FOXP3(+) cells present in the CD4(+)CD25(low) subset. Flow cytometric analysis of PB identified five-fold more Treg, determined by FOXP3 expression, in the CD4(+)CD25(+)TNFR2(+) subset than in the CD4(+)CD25(high) subset. In addition, similar levels of FOXP3(+) cells were identified in both the CD4(+)CD25(+)TNFR2(+) and CD4(+)CD25(+)CD127(low/-) subsets. Furthermore, the CD4(+)CD25(+)TNFR2(+) subset expressed high levels of CTLA-4, CD45RO, CCR4 and low levels of CD45RA and CD127, a phenotype characteristic of Treg. Upon TCR stimulation, human PB CD4(+)CD25(+)TNFR2(+) cells were anergic and markedly inhibited the proliferation and cytokine production of co-cultured T-responder cells. In contrast, CD4(+)CD25(+)TNFR2(-) and CD4(+)CD25(-)TNFR2(+) T cells did not show inhibitory activity. As some non-Treg express TNFR2, the combination of CD25 and TNFR2 must be used to identify a larger population of human Treg, a population that may prove to be of diagnostic and therapeutic benefit in cancer and autoimmune diseases.


Subject(s)
Forkhead Transcription Factors/analysis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/classification , Adult , Antigen Presentation , Antigens, CD/analysis , CTLA-4 Antigen , Cells, Cultured/immunology , Cells, Cultured/metabolism , Coculture Techniques , Flow Cytometry , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-7 Receptor alpha Subunit/analysis , Leukocyte Common Antigens/analysis , Lymphocyte Activation/drug effects , Receptors, Antigen, T-Cell/immunology , Receptors, Tumor Necrosis Factor, Type II/analysis , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
J Immunol ; 180(10): 6467-71, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18453563

ABSTRACT

TNFR2 is predominantly expressed by a subset of human and mouse CD4(+)CD25(+)FoxP3(+) T regulatory cells (Tregs). In this study, we characterized the phenotype and function of TNFR2(+) Tregs in peripheral lymphoid tissues of normal and tumor-bearing C57BL/6 mice. We found that TNFR2 was expressed on 30-40% of the Tregs of the peripheral activated/memory subset that were most highly suppressive. In contrast, TNFR2(-) Tregs exhibited the phenotype of naive cells and only had minimal suppressive activity. Although not typically considered to be Tregs, CD4(+)CD25(-)TNFR2(+) cells nevertheless possessed moderate suppressive activity. Strikingly, the suppressive activity of TNFR2(+) Tregs was considerably more potent than that of reportedly highly suppressive CD103(+) Tregs. In the Lewis lung carcinoma model, more highly suppressive TNFR2(+) Tregs accumulated intratumorally than in the periphery. Thus, TNFR2 identifies a unique subset of mouse Tregs with an activated/memory phenotype and maximal suppressive activity that may account for tumor-infiltrating lymphocyte-mediated immune evasion by tumors.


Subject(s)
Carcinoma, Lewis Lung/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Antigens/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/immunology
6.
Curr Protoc Immunol ; Chapter 10: Unit 10.29, 2003 Aug.
Article in English | MEDLINE | ID: mdl-18432896

ABSTRACT

The multiprobe RNase protection assay enables investigators to monitor RNA expression of 8-12 genes with as little as 1 microg of total RNA. The commercial availability of numerous multi-gene template sets makes this assay practical for all basic research programs.


Subject(s)
Genetic Techniques , Molecular Probe Techniques , Molecular Probes , RNA, Messenger/analysis , Ribonucleases/genetics , Animals , Humans , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...