Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 46(1): 78-91, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099866

ABSTRACT

FoxP3-expressing regulatory T (Treg) cells are essential for maintaining immune homeostasis. Activated Treg cells undergo further differentiation into an effector state that highly expresses genes critical for Treg cell function, although how this process is coordinated on a transcriptional level is poorly understood. Here, we demonstrate that mice lacking the transcription factor Myb in Treg cells succumbed to a multi-organ inflammatory disease. Myb was specifically expressed in, and required for the differentiation of, thymus-derived effector Treg cells. The combination of transcriptome and genomic footprint analyses revealed that Myb directly regulated a large proportion of the gene expression specific to effector Treg cells, identifying Myb as a critical component of the gene regulatory network controlling effector Treg cell differentiation and function.


Subject(s)
Gene Regulatory Networks/immunology , Homeostasis/immunology , Lymphocyte Activation/immunology , Proto-Oncogene Proteins c-myb/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcriptome
2.
Nat Immunol ; 12(4): 304-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21378976

ABSTRACT

Regulatory T cells (T(reg) cells) are required for peripheral tolerance. Evidence indicates that T(reg) cells can adopt specialized differentiation programs in the periphery that are controlled by transcription factors usually associated with helper T cell differentiation. Here we demonstrate that expression of the transcription factor Blimp-1 defined a population of T(reg) cells that localized mainly to mucosal sites and produced IL-10. Blimp-1 was required for IL-10 production by these cells and for their tissue homeostasis. We provide evidence that the transcription factor IRF4, but not the transcription factor T-bet, was essential for Blimp-1 expression and for the differentiation of all effector T(reg) cells. Thus, our study defines a differentiation pathway that leads to the acquisition of T(reg) cell effector functions and requires both IRF4 and Blimp-1.


Subject(s)
Cell Differentiation/genetics , Interferon Regulatory Factors/genetics , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Positive Regulatory Domain I-Binding Factor 1 , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes, Regulatory/cytology , Transcription Factors/metabolism
3.
PLoS Pathog ; 12(1): e1005398, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26765224

ABSTRACT

Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation.


Subject(s)
Inflammation/immunology , Interleukin-10/biosynthesis , Leishmaniasis, Visceral/immunology , Repressor Proteins/immunology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Inflammation/pathology , Interleukin-10/immunology , Leishmaniasis, Visceral/pathology , Malaria/immunology , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes, Regulatory/immunology
4.
J Autoimmun ; 91: 73-82, 2018 07.
Article in English | MEDLINE | ID: mdl-29724515

ABSTRACT

Regulatory T (Treg) cells maintain immunological tolerance in steady-state and after immune challenge. Activated Treg cells can undergo further differentiation into an effector state that highly express genes critical for Treg cell function, including ICOS, TIGIT and IL-10, although how this process is controlled is poorly understood. Effector Treg cells also specifically express the transcriptional regulator Blimp-1 whose expression overlaps with many of the canonical markers associated with effector Treg cells, although not all ICOS+TIGIT+ Treg cells express Blimp-1 or IL-10. In this study, we addressed the role of Blimp-1 in effector Treg cell function. Mice lacking Blimp-1 specifically in Treg cells mature normally, but succumb to a multi-organ inflammatory disease later in life. Blimp-1 is not required for Treg cell differentiation, with mutant mice having increased numbers of effector Treg cells, but regulated a suite of genes involved in cell signaling, communication and survival, as well as being essential for the expression of the immune modulatory cytokine IL-10. Thus, Blimp-1 is a marker of effector Treg cells in all contexts examined and is required for the full functionality of these cells during aging.


Subject(s)
Aging/immunology , Inflammation/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immune Tolerance , Inflammation/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/genetics , Signal Transduction
5.
Immunity ; 30(1): 56-66, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19119023

ABSTRACT

Apoptotic death of hepatocytes, a contributor to many chronic and acute liver diseases, can be a consequence of overactivation of the immune system and is often mediated by TNFalpha. Injection with lipopolysaccharide (LPS) plus the transcriptional inhibitor D(+)-galactosamine (GalN) or mitogenic T cell activation causes fatal hepatocyte apoptosis in mice, which is mediated by TNFalpha, but the effector mechanisms remain unclear. Our analysis of gene-targeted mice showed that caspase-8 is essential for hepatocyte killing in both settings. Loss of Bid, the proapoptotic BH3-only protein activated by caspase-8 and essential for Fas ligand-induced hepatocyte killing, resulted only in a minor reduction of liver damage. However, combined loss of Bid and another BH3-only protein, Bim, activated by c-Jun N-terminal kinase (JNK), protected mice from LPS+GalN-induced hepatitis. These observations identify caspase-8 and the BH3-only proteins Bid and Bim as potential therapeutic targets for treatment of inflammatory liver diseases.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 8/metabolism , Chemical and Drug Induced Liver Injury , Hepatocytes/pathology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bcl-2-Like Protein 11 , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism
6.
Cancer Immunol Immunother ; 66(4): 515-522, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28116480

ABSTRACT

Analysis of tumour-infiltrating T cells in colorectal cancer can predict disease-free survival. The Immunoscore, obtained by quantifying tumour-infiltrating CD3+ and CD8+ T cells, may improve current staging. Effector regulatory T cells are a potently suppressive subset in mice and, while present in human colorectal cancer, their role in patient outcome is unknown. Immunofluorescence was used to analyse immune cell infiltrates in patients with early (stage II) colorectal cancer with (n = 13) and without (n = 19) recurrent disease. CD3 and CD8 were used for the Immunoscore; FOXP3, BLIMP-1 and CD3 to identify effector regulatory T cells. Patients with high Immunoscores had increased disease-free survival compared to patients with low Immunoscores (Log-rank test p < 0.01). Prediction of outcome was further improved by stratifying patients with a low Immunoscore according to CD3+FOXP3+BLIMP-1+ cell infiltration at the invasive margin. Patients with a low Immunoscore and high infiltrate of CD3+FOXP3+BLIMP-1+ cells tended to have better disease-free survival than patients with low Immunoscore and low infiltrate of CD3+FOXP3+BLIMP-1+ cells. Patients with a high Immunoscore had better disease-free survival than patients with a low Immunoscore and low infiltrate of CD3+ FOXP3+ BLIMP-1+ cells (Log-rank test p < 0.001). These results indicate that tumour infiltration with effector regulatory T cells improves the prognostic value of the Immunoscore and implies that these cells may play a role in colorectal cancer patient outcome.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/diagnosis , Lymphocytes, Tumor-Infiltrating/immunology , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Aged , Aged, 80 and over , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Forkhead Transcription Factors/metabolism , Humans , Immunologic Tests , Male , Middle Aged , Neoplasm Staging , New Zealand , Pilot Projects , Positive Regulatory Domain I-Binding Factor 1 , Predictive Value of Tests , Prognosis , Survival Analysis
7.
Immunol Cell Biol ; 94(8): 796-801, 2016 09.
Article in English | MEDLINE | ID: mdl-27140932

ABSTRACT

The transcriptional repressor/activator interferon regulatory factor 8 (IRF8) modulates the differentiation of a multitude of hematopoietic lineages. However, the role of IRF8 in CD4(+) T-cell development is less well defined, with a recent study implicating IRF8 as an intrinsic repressor of interleukin-17 (IL-17) expressing T helper type 17 (Th17) cell differentiation. Using an IRF8-EGFP reporter strain we have confirmed that IRF8 is expressed in all T helper lineages, including Th17 cells. The loss of IRF8 did not affect Th17 differentiation in vitro, beyond a small increase in IL-22 expression. Moreover, IRF8 deficiency did not enhance the Th17 immune response in experimental T-cell transfer colitis. Together, these results suggest that IRF8 does not play an essential intrinsic role in Th17 cell differentiation.


Subject(s)
Cell Differentiation , Interferon Regulatory Factors/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Colitis/immunology , Colitis/pathology , Lymphocyte Activation/immunology , Mice , Mice, Knockout
8.
Trends Immunol ; 34(2): 74-80, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219401

ABSTRACT

Regulatory T (Treg) cells are essential for immunological tolerance and homeostasis. Although forkhead box (Fox)p3 is continually required to reinforce the Treg cell program, Treg cells can also undergo stimulus-specific differentiation that is regulated by transcription factors typically associated with the differentiation of conventional CD4(+) T cells. This results in effector Treg (eTreg) cells with unique migratory and functional properties matched to the stimulus that elicited the initial response. Despite this functional and transcriptional heterogeneity, expression of the transcription factor B lymphocyte-induced maturation protein (Blimp)-1, a key player in late B cell and conventional T cell differentiation, is common to all eTreg cells. Here, we discuss the factors that control the differentiation of eTreg cells and their importance in disease settings.


Subject(s)
Autoimmune Diseases/immunology , Cell Differentiation , Communicable Diseases/immunology , Forkhead Transcription Factors/metabolism , Neoplasms/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/genetics , Humans , Mice , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism
10.
J Exp Med ; 202(5): 583-8, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16129707

ABSTRACT

The activation NKG2D receptor has been shown to play an important role in the control of experimental tumor growth and metastases expressing ligands for NKG2D; however, a function for this recognition pathway in host protection from de novo tumorigenesis has never been demonstrated. We show that neutralization of NKG2D enhances the sensitivity of wild-type (WT) C57BL/6 and BALB/c mice to methylcholanthrene (MCA)-induced fibrosarcoma. The importance of the NKG2D pathway was additionally illustrated in mice deficient for either IFN-gamma or tumor necrosis factor-related apoptosis-inducing ligand, whereas mice depleted of natural killer cells, T cells, or deficient for perforin did not display any detectable NKG2D phenotype. Furthermore, IL-12 therapy preventing MCA-induced sarcoma formation was also largely dependent on the NKG2D pathway. Although NKG2D ligand expression was variable or absent on sarcomas emerging in WT mice, sarcomas derived from perforin-deficient mice were Rae-1(+) and immunogenic when transferred into WT syngeneic mice. These findings suggest an important early role for the NKG2D in controlling and shaping tumor formation.


Subject(s)
Fibrosarcoma/immunology , Fibrosarcoma/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Animals , Antibodies, Monoclonal , Apoptosis Regulatory Proteins/deficiency , Carrier Proteins/metabolism , Fibrosarcoma/chemically induced , Fibrosarcoma/drug therapy , Flow Cytometry , Histocompatibility Antigens Class I/metabolism , Interleukin-12/metabolism , Interleukin-12/therapeutic use , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Glycoproteins/deficiency , Membrane Proteins , Methylcholanthrene , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K , Neutralization Tests , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Natural Killer Cell
11.
Proc Natl Acad Sci U S A ; 105(31): 10895-900, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18667695

ABSTRACT

Chronic cholestasis often results in premature death from liver failure with fibrosis; however, the molecular mechanisms contributing to biliary cirrhosis are not demonstrated. In this article, we show that the death signal mediated by TNF-related apoptosis-inducing ligand (TRAIL) receptor 2/death receptor 5 (DR5) may be a key regulator of cholestatic liver injury. Agonistic anti-DR5 monoclonal antibody treatment triggered cholangiocyte apoptosis, and subsequently induced cholangitis and cholestatic liver injury in a mouse strain-specific manner. TRAIL- or DR5-deficient mice were relatively resistant to common bile duct ligation-induced cholestasis, and common bile duct ligation augmented DR5 expression on cholangiocytes, sensitizing mice to DR5-mediated cholangitis. Notably, anti-DR5 monoclonal antibody-induced cholangitis exhibited the typical histological appearance, reminiscent of human primary sclerosing cholangitis. Human cholangiocytes constitutively expressed DR5, and TRAIL expression and apoptosis were significantly elevated in cholangiocytes of human primary sclerosing cholangitis and primary biliary cirrhosis patients. Thus, TRAIL/DR5-mediated apoptosis may substantially contribute to chronic cholestatic disease, particularly primary sclerosing cholangitis.


Subject(s)
Apoptosis/immunology , Cholangitis/metabolism , Cholestasis/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Cholangitis/pathology , Cholestasis/metabolism , Cholestasis/pathology , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Mutant Strains , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
12.
J Exp Med ; 195(2): 161-9, 2002 Jan 21.
Article in English | MEDLINE | ID: mdl-11805143

ABSTRACT

Natural killer (NK) cells and interferon (IFN)-gamma have been implicated in immune surveillance against tumor development. Here we show that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays a critical role in the NK cell-mediated and IFN-gamma-dependent tumor surveillance. Administration of neutralizing monoclonal antibody against TRAIL promoted tumor development in mice subcutaneously inoculated with a chemical carcinogen methylcholanthrene (MCA). This protective effect of TRAIL was at least partly mediated by NK cells and totally dependent on IFN-gamma. In the absence of TRAIL, NK cells, or IFN-gamma, TRAIL-sensitive sarcomas preferentially emerged in MCA-inoculated mice. Moreover, development of spontaneous tumors in p53(+/-) mice was also promoted by neutralization of TRAIL. These results indicated a substantial role of TRAIL as an effector molecule that eliminates developing tumors.


Subject(s)
Cytotoxicity, Immunologic/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Membrane Glycoproteins/immunology , Neoplasms, Experimental/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , Cytotoxicity, Immunologic/genetics , Interferon-gamma/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/prevention & control , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
13.
J Exp Med ; 200(10): 1325-35, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15545356

ABSTRACT

Single and combination cytokines offer promise in some patients with advanced cancer. Many spontaneous and experimental cancers naturally express ligands for the lectin-like type-2 transmembrane stimulatory NKG2D immunoreceptor; however, the role this tumor recognition pathway plays in immunotherapy has not been explored to date. Here, we show that natural expression of NKG2D ligands on tumors provides an effective target for some cytokine-stimulated NK cells to recognize and suppress tumor metastases. In particular, interleukin (IL)-2 or IL-12 suppressed tumor metastases largely via NKG2D ligand recognition and perforin-mediated cytotoxicity. By contrast, IL-18 required tumor sensitivity to Fas ligand (FasL) and surprisingly did not depend on the NKG2D-NKG2D ligand pathway. A combination of IL-2 and IL-18 stimulated both perforin and FasL effector mechanisms with very potent effects. Cytokines that stimulated perforin-mediated cytotoxicity appeared relatively more effective against tumor metastases expressing NKG2D ligands. These findings indicate that a rational choice of cytokines can be made given the known sensitivity of tumor cells to perforin, FasL, and tumor necrosis factor-related apoptosis-inducing ligand and the NKG2D ligand status of tumor metastases.


Subject(s)
Cytokines/therapeutic use , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasm Metastasis/therapy , Receptors, Immunologic/metabolism , Animals , Apoptosis/immunology , Cytokines/immunology , Cytokines/metabolism , Fas Ligand Protein , Flow Cytometry , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Ligands , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K , Pore Forming Cytotoxic Proteins , Receptors, Natural Killer Cell , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured
14.
J Exp Med ; 198(3): 491-6, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12900523

ABSTRACT

The molecular basis of thymocyte negative selection, which plays a critical role in establishing and maintaining immunological tolerance, is not yet resolved. In particular, the importance of the death receptor subgroup of the tumor necrosis factor (TNF)-family has been the subject of many investigations, with equivocal results. A recent report suggested that TRAIL was a critical factor in this process, a result that does not fit well with previous studies that excluded a role for the FADD-caspase 8 pathway, which is essential for TRAIL and Fas ligand (FasL) signaling, in negative selection. We have investigated intrathymic negative selection of TRAIL-deficient thymocytes, using four well-established models, including antibody-mediated TCR/CD3 ligation in vitro, stimulation with endogenous superantigen in vitro and in vivo, and treatment with exogenous superantigen in vitro. We were unable to demonstrate a role for TRAIL signaling in any of these models, suggesting that this pathway is not a critical factor for thymocyte negative selection.


Subject(s)
Clonal Deletion , Membrane Glycoproteins/metabolism , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigens, CD/metabolism , Apoptosis Regulatory Proteins , Gene Deletion , Lymph Nodes/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Spleen/cytology , T-Lymphocyte Subsets , T-Lymphocytes/cytology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics
15.
J Exp Med ; 200(6): 811-6, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15365097

ABSTRACT

About 30% of cases of the autosomal recessive immunodeficiency disorder hemophagocytic lymphohistiocytosis are believed to be caused by inactivating mutations of the perforin gene. We expressed perforin in rat basophil leukemia cells to define the basis of perforin dysfunction associated with two mutations, R225W and G429E, inherited by a compound heterozygote patient. Whereas RBL cells expressing wild-type perforin (67 kD) efficiently killed Jurkat target cells to which they were conjugated, the substitution to tryptophan at position 225 resulted in expression of a truncated ( approximately 45 kD) form of the protein, complete loss of cytotoxicity, and failure to traffic to rat basophil leukemia secretory granules. By contrast, G429E perforin was correctly processed, stored, and released, but the rat basophil leukemia cells possessed reduced cytotoxicity. The defective function of G429E perforin mapped downstream of exocytosis and was due to its reduced ability to bind lipid membranes in a calcium-dependent manner. This study elucidates the cellular basis for perforin dysfunctions in hemophagocytic lymphohistiocytosis and provides the means for studying structure-function relationships for lymphocyte perforin.


Subject(s)
Histiocytosis, Non-Langerhans-Cell/genetics , Membrane Glycoproteins/genetics , Mutation, Missense , Animals , Histiocytosis, Non-Langerhans-Cell/immunology , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Perforin , Pore Forming Cytotoxic Proteins , Rats , T-Lymphocytes, Cytotoxic/immunology
16.
J Leukoc Biol ; 85(1): 108-16, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18948547

ABSTRACT

The generalized lymphoproliferative disorder (gld) mouse strain is characterized by severe splenomegaly/lymphadenopathy, the production of autoimmune antibodies, and the appearance of CD4/CD8-negative T cells. An additional TNF deficiency of gld/gld mice attenuates the course of the disorder through a yet-unknown mechanism. In this study, we could demonstrate that the reduced splenomegaly and lymphadenopathy in B6.gld/gld.TNF-/- mice were correlated with a decreased peripheral T cell proliferation rate and a delayed polyclonal activation. A comparative analysis of naïve T cells and memory/effector T cells showed an age-dependent difference in the T cell activation pattern in the spleen of B6.gld/gld and B6.gld/gld.TNF-/- mice. T cells from B6.gld/gld.TNF-/- spleens and lymph nodes showed significantly higher levels of CCR7 and CD62 ligand on their surface compared with B6.gld/gld mice when mice of the same age were compared. Additionally, we found an increased titer of the Th1 cytokine IFN-gamma in the serum of B6.gld/gld mice, whereas the concentration of IFN-gamma was markedly reduced in the serum of B6.gld/gld.TNF-/- mice. These findings support the hypothesis that increased T cell activation and proliferation in the presence of TNF contribute to the exacerbation of the gld syndrome.


Subject(s)
Aging/metabolism , Lymphocyte Activation , Lymphoproliferative Disorders/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Differentiation , Cell Proliferation , Hyaluronan Receptors/metabolism , Interferon-gamma/blood , L-Selectin/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Diseases/immunology , Lymphatic Diseases/metabolism , Lymphatic Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR7/metabolism , Spleen/immunology , Spleen/pathology , Splenomegaly/immunology , Splenomegaly/metabolism , Splenomegaly/pathology , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/genetics
17.
Int Immunol ; 20(2): 267-76, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18192669

ABSTRACT

The contribution of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway to intrathymic negative selection is a controversial subject with two studies suggesting a key role for TRAIL, while others demonstrated normal negative selection, in TRAIL- and TRAIL receptor-deficient mice. The basis of these discrepancies is unclear and may in part reflect differences in the negative selection models under investigation. Considering the importance of the negative selection process in the establishment of a competent immune system, it is essential that these discrepancies be fully resolved. In this study, we failed to identify a role for TRAIL in an acute model of peptide antigen-specific negative selection using a TCR transgenic system as well as antibody-mediated TCR/CD3 ligation in vitro and in vivo. Moreover, thymic dendritic cells, the main cellular mediators of negative selection in the thymus, did not constitutively express TRAIL, and TRAIL receptor (DR5) expression was negative or extremely low on thymocytes. Furthermore, in vitro thymocyte deletion was normal in C57BL/6 TRAIL(-/-) gld mice, suggesting that TRAIL and FasL do not function cooperatively to induce negative selection. These results, combined with the fact that aged C57BL/6 TRAIL(-/-) mice showed no signs of spontaneous autoimmunity, strongly indicate that intrathymic negative selection occurs normally in the absence of TRAIL signaling.


Subject(s)
Selection, Genetic , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Apoptosis , Autoimmune Diseases , CD3 Complex , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/deficiency , TNF-Related Apoptosis-Inducing Ligand/genetics , Thymus Gland/cytology
18.
JAMA Neurol ; 75(6): 681-689, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29507931

ABSTRACT

Importance: Neuroinflammation appears to be a key modulator of disease progression in amyotrophic lateral sclerosis (ALS) and thereby a promising therapeutic target. The CD4+Foxp3+ regulatory T-cells (Tregs) infiltrating into the central nervous system suppress neuroinflammation and promote the activation of neuroprotective microglia in mouse models of ALS. To our knowledge, the therapeutic association of host Treg expansion with ALS progression has not been studied in vivo. Objective: To assess the role of Tregs in regulating the pathophysiology of ALS in humans and the therapeutic outcome of increasing Treg activity in a mouse model of the disease. Design, Setting, and Participants: This prospective multicenter human and animal study was performed in hospitals, outpatient clinics, and research institutes. Clinical and function assessment, as well as immunological studies, were undertaken in 33 patients with sporadic ALS, and results were compared with 38 healthy control participants who were consecutively recruited from the multidisciplinary ALS clinic at Westmead Hospital between February 1, 2013, and December 31, 2014. All data analysis on patients with ALS was undertaken between January 2015 and December 2016. Subsequently, we implemented a novel approach to amplify the endogenous Treg population using peripheral injections of interleukin 2/interleukin 2 monoclonal antibody complexes (IL-2c) in transgenic mice that expressed mutant superoxide dismutase 1 (SOD1), a gene associated with motor neuron degeneration. Main Outcomes and Measures: In patients with ALS, Treg levels were determined and then correlated with disease progression. Circulating T-cell populations, motor neuron size, glial cell activation, and T-cell and microglial gene expression in spinal cords were determined in SOD1G93A mice, as well as the association of Treg amplification with disease onset and survival time in mice. Results: The cohort of patients with ALS included 24 male patients and 9 female patients (mean [SD] age at assessment, 58.9 [10.9] years). There was an inverse correlation between total Treg levels (including the effector CD45RO+ subset) and rate of disease progression (R = -0.40, P = .002). Expansion of the effector Treg population in the SOD1G93A mice was associated with a significant slowing of disease progression, which was accompanied by an increase in survival time (IL-2c-treated mice: mean [SD], 160.6 [10.8] days; control mice: mean [SD], 144.9 [10.6] days; P = .003). Importantly, Treg expansion was associated with preserved motor neuron soma size and marked suppression of astrocytic and microglial immunoreactivity in the spinal cords of SOD1G93A mice, as well as elevated neurotrophic factor gene expression in spinal cord and peripheral nerves. Conclusions and Relevance: These findings establish a neuroprotective effect of Tregs, possibly mediated by suppression of toxic neuroinflammation in the central nervous system. Strategies aimed at enhancing the Treg population and neuroprotective activity from the periphery may prove therapeutically useful for patients with ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/genetics , Disease Models, Animal , Disease Progression , T-Lymphocytes, Regulatory/metabolism , Aged , Amyotrophic Lateral Sclerosis/diagnosis , Animals , Cohort Studies , Cross-Sectional Studies , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Prospective Studies , Superoxide Dismutase/genetics
19.
Int J Biochem Cell Biol ; 39(2): 280-6, 2007.
Article in English | MEDLINE | ID: mdl-17097329

ABSTRACT

Cancer is a widespread disease, with half of all men and one-third of all women in the United States developing cancer during their lifetime. The efficacy of many cancer treatments including radiotherapy, chemotherapy and immunotherapy is due to their ability to induce tumor cell apoptosis. Recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is currently being developed as a cancer therapeutic since it selectively induces apoptosis in a variety of transformed cells, but not in most normal cells. Agonistic monoclonal antibodies (mAbs) specific for human death-inducing TRAIL receptors (DR4 or DR5) are also being actively pursued. Importantly, in experimental mice, synergistic anti-tumor effects have been observed with a combination treatment of agonistic mAb against DR5 together with either IL-21 or agonistic mAbs against CD40 and CD137. Together, these findings suggest that antibody-based therapies that cause tumor cell apoptosis and promote T cell memory or function may be effective in fighting cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal/immunology , Clinical Trials as Topic , Drug Synergism , Humans , Interleukins/therapeutic use , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
20.
Mol Immunol ; 42(4): 501-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15607806

ABSTRACT

Natural killer (NK) cells are innate effector lymphocytes necessary for defence against stressed, microbe-infected, or malignant cells. NK cells kill target cells by either of two major mechanisms that require direct contact between NK cells and target cells. In the first pathway, cytoplasmic granule toxins, predominantly a membrane-disrupting protein known as perforin, and a family of structurally related serine proteases (granzymes) with various substrate specificities, are secreted by exocytosis and together induce apoptosis of the target cell. The granule-exocytosis pathway potently activates cell-death mechanisms that operate through the activation of apoptotic cysteine proteases (caspases), but can also cause cell death in the absence of activated caspases. The second pathway involves the engagement of death receptors (e.g. Fas/CD95) on target cells by their cognate ligands (e.g. FasL) on NK cells, resulting in classical caspase-dependent apoptosis. The comparative role of these pathways in the pathophysiology of many diseases is being dissected by analyses of gene-targeted mice that lack these molecules, and humans who have genetic mutations affecting these pathways. We are also now learning that the effector function of NK cells is controlled by interactions involving specific NK cell receptors and their cognate ligands, either on target cells, or other cells of the immune system. This review will discuss the functional importance of NK cell cytotoxicity and the receptor/ligand interactions that control these processes.


Subject(s)
Cytotoxicity, Immunologic/physiology , Killer Cells, Natural/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/physiology , Apoptosis , Cytokines/immunology , Cytoplasmic Granules/physiology , Exocytosis/physiology , Humans , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/physiology , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Tumor Necrosis Factor/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL