Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Publication year range
1.
Nature ; 527(7577): 249-53, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26503055

ABSTRACT

Epigenetic silencing including histone modifications and DNA methylation is an important tumorigenic mechanism. However, its role in cancer immunopathology and immunotherapy is poorly understood. Using human ovarian cancers as our model, here we show that enhancer of zeste homologue 2 (EZH2)-mediated histone H3 lysine 27 trimethylation (H3K27me3) and DNA methyltransferase 1 (DNMT1)-mediated DNA methylation repress the tumour production of T helper 1 (TH1)-type chemokines CXCL9 and CXCL10, and subsequently determine effector T-cell trafficking to the tumour microenvironment. Treatment with epigenetic modulators removes the repression and increases effector T-cell tumour infiltration, slows down tumour progression, and improves the therapeutic efficacy of programmed death-ligand 1 (PD-L1; also known as B7-H1) checkpoint blockade and adoptive T-cell transfusion in tumour-bearing mice. Moreover, tumour EZH2 and DNMT1 are negatively associated with tumour-infiltrating CD8(+) T cells and patient outcome. Thus, epigenetic silencing of TH1-type chemokines is a novel immune-evasion mechanism of tumours. Selective epigenetic reprogramming alters the T-cell landscape in cancer and may enhance the clinical efficacy of cancer therapy.


Subject(s)
Chemokines/genetics , Epigenesis, Genetic , Gene Silencing , Immunotherapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Th1 Cells/metabolism , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokines/biosynthesis , Chemokines/immunology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/drug effects , Female , Histones/chemistry , Histones/metabolism , Humans , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lysine/metabolism , Mice , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Prognosis , Th1 Cells/immunology , Tumor Cells, Cultured , Tumor Escape/immunology , Xenograft Model Antitumor Assays
2.
J Neurovirol ; 25(6): 844-852, 2019 12.
Article in English | MEDLINE | ID: mdl-31278536

ABSTRACT

In this article, we studied the production of the chemokine CXCL9, also termed Mig (monokine induced by gamma interferon) by cultured SJL/J mouse astrocytes infected with the BeAn strain of Theiler's murine encephalomyelitis virus (TMEV). This picornavirus induces demyelination in the SJL/J genetically susceptible strain of mice through an immune process mediated by CD4+ Th1 T cells. Those cells were chemoattracted by chemokines inside the central nervous system (CNS) after blood-brain barrier (BBB) disruption.cRNAs from TMEV- and mock-infected astrocytes cells were hybridized to the Affymetrix murine genome U74v2 DNA microarray. Hybridization data analysis revealed the upregulation of six sequences potentially coding for Mig. We confirmed post infection Mig mRNA increase by quantitative (qPCR) and RT-PCR. The presence of Mig in the supernatants of infected astrocytes was quantified using a specific ELISA. Secreted Mig was biologically active, inducing chemoattraction of mouse activated CD4+ T lymphocytes. Conversely, attracting activity on CD3+ resting T cells that can be attributed to chemokines as CXCL12/SDF-1α could not be demonstrated in these supernatants. No overinduction of the gene coding for this chemokine was assessed by DNA hybridization either. Both recombinant IFN-γ and TNF-α inflammatory cytokines were also strong inducers of Mig in SJL/J astrocyte cultures.


Subject(s)
Astrocytes/immunology , Astrocytes/virology , Cardiovirus Infections/immunology , Chemokine CXCL9/immunology , Th1 Cells/immunology , Animals , Chemokine CXCL9/biosynthesis , Chemotaxis, Leukocyte/immunology , Lymphocyte Activation/immunology , Mice , Theilovirus
3.
PLoS Pathog ; 11(12): e1005319, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646682

ABSTRACT

The innate immune system relies to a great deal on the interaction of pattern recognition receptors with pathogen- or damage-associated molecular pattern molecules. Extracellular histones belong to the latter group and their release has been described to contribute to the induction of systemic inflammatory reactions. However, little is known about their functions in the early immune response to an invading pathogen. Here we show that extracellular histones specifically target monocytes in human blood and this evokes the mobilization of the chemotactic chemokines CXCL9 and CXCL10 from these cells. The chemokine induction involves the toll-like receptor 4/myeloid differentiation factor 2 complex on monocytes, and is under the control of interferon-γ. Consequently, subcutaneous challenge with extracellular histones results in elevated levels of CXCL10 in a murine air pouch model and an influx of leukocytes to the site of injection in a TLR4 dependent manner. When analyzing tissue biopsies from patients with necrotizing fasciitis caused by Streptococcus pyogenes, extracellular histone H4 and CXCL10 are immunostained in necrotic, but not healthy tissue. Collectively, these results show for the first time that extracellular histones have an important function as chemoattractants as their local release triggers the recruitment of immune cells to the site of infection.


Subject(s)
Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Chemotaxis, Leukocyte/immunology , Histones/immunology , Leukocytes/immunology , Animals , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Chemokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Leukocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Monocytes/immunology , Surface Plasmon Resonance
4.
Br J Cancer ; 114(6): 697-703, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26889977

ABSTRACT

BACKGROUND: The novel chemokine CXCL17 acts as chemoattractant for monocytes, macrophages and dendritic cells. CXCL17 also has a role in angiogenesis of importance for tumour development. METHODS: Expression of CXCL17, CXCL10, CXCL9 and CCL2 was assessed in primary colon cancer tumours, colon carcinoma cell lines and normal colon tissue at mRNA and protein levels by real-time qRT-PCR, immunohistochemistry, two-colour immunofluorescence and immunomorphometry. RESULTS: CXCL17 mRNA was expressed at 8000 times higher levels in primary tumours than in normal colon (P < 0.0001). CXCL17 protein was seen in 17.2% of cells in tumours as compared with 0.07% in normal colon (P = 0.0002). CXCL10, CXCL9 and CCL2 mRNAs were elevated in tumours but did not reach the levels of CXCL17. CXCL17 and CCL2 mRNA levels were significantly correlated in tumours. Concordant with the mRNA results, CXCL10- and CXCL9-positive cells were detected in tumour tissue, but at significantly lower numbers than CXCL17. Two-colour immunofluorescence and single-colour staining of consecutive sections for CXCL17 and the epithelial cell markers carcinoembryonic antigen and BerEP4 demonstrated that colon carcinoma tumour cells indeed expressed CXCL17. CONCLUSIONS: CXCL17 is ectopically expressed in primary colon cancer tumours. As CXCL17 enhances angiogenesis and attracts immune cells, its expression could be informative for prognosis in colon cancer patients.


Subject(s)
Chemokines/biosynthesis , Colonic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Caco-2 Cells , Cell Line, Tumor , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Chemokines/genetics , Chemokines, CXC , Colonic Neoplasms/genetics , Female , Fluorescent Antibody Technique , HT29 Cells , Humans , Immunohistochemistry , Male , Middle Aged , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
5.
Eur J Immunol ; 45(4): 1059-1068, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25616060

ABSTRACT

Bcl-3 is an atypical member of the IκB family. Bcl-3 functions as a cofactor of p50/NF-κB1 or p52/NF-κB2 homodimers in nuclei, where it modulates NF-κB-regulated transcription in a context-dependent way. Bcl-3 has tumorigenic potential, is critical in host defense of pathogens, and has been reported to ameliorate or exacerbate inflammation, depending on disease model. However, cell-specific functions of Bcl-3 remain largely unknown. Here, we explored the role of Bcl-3 in a contact hypersensitivity (CHS) mouse model, which depends on the interplay between keratinocytes and immune cells. Bcl-3-deficient mice exhibited an exacerbated and prolonged CHS response to oxazolone. Increased inflammation correlated with higher production of chemokines CXCL2, CXCL9, and CXCL10, and consequently increased recruitment of neutrophils and CD8(+) T cells. BM chimera experiments indicated that the ability of Bcl-3 to reduce the CHS response depended on Bcl-3 activity in radioresistant cells. Specific ablation of Bcl-3 in keratinocytes resulted in increased production of CXCL9 and CXCL10 and sustained recruitment of specifically CD8(+) T cells. These findings identify Bcl-3 as a critical player during the later stage of the CHS reaction to limit inflammation via actions in radioresistant cells, including keratinocytes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dermatitis, Allergic Contact/immunology , Inflammation/immunology , Neutrophils/immunology , Proto-Oncogene Proteins/metabolism , Radiation Tolerance/immunology , Transcription Factors/metabolism , Animals , B-Cell Lymphoma 3 Protein , Chemokine CXCL10/biosynthesis , Chemokine CXCL2/biosynthesis , Chemokine CXCL9/biosynthesis , Inflammation/chemically induced , Inflammation Mediators , Keratinocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Oxazolone , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
6.
Eur J Immunol ; 45(4): 971-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25707546

ABSTRACT

B-cell lymphoma (Bcl)-3 is a nonclassical member of the IκB protein family known to interact with transcriptionally inactive NF-κB1 and NF-κB2 homodimers to modulate gene expression. Besides its action as an oncoprotein, Bcl-3 has been shown to have both proinflammatory and anti-inflammatory functions depending on the cell-type affected. In this issue of the European Journal of Immunology, Tassi et al. [Eur. J. Immunol. 2015. 45: 1059-1068] report that Bcl-3 inhibits the production of the proinflammatory chemokines CXCL9 and CXCL10 in keratinocytes, thereby restricting the influx of CD8(+) effector T cells in a mouse model of allergic contact dermatitis. In addition, mice with a global deficiency of Bcl-3 show enhanced ear swelling responses in the late phase of contact hypersensitivity responses. Besides keratinocytes, other radioresistant cell types appear to also utilize Bcl-3 to dampen the inflammatory response. This Commentary will discuss the evidence supporting Bcl-3 as a critical player in limiting inflammation during the later stages of contact hypersensitivity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Dermatitis, Allergic Contact/immunology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , B-Cell Lymphoma 3 Protein , Ear/pathology , Inflammation/immunology , Keratinocytes/metabolism , Mice , Mice, Inbred NOD , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics
7.
J Immunol ; 192(4): 1671-80, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442435

ABSTRACT

Ribavirin has proven to be a key component of hepatitis C therapies both involving IFNs and new direct-acting antivirals. The hepatitis C virus-mediated interference with intrahepatic immunity by cleavage of mitochondrial antiviral signaling protein (MAVS) and T cell protein tyrosine phosphatase (TCPTP) suggests an avenue for compounds that may counteract these effects. We therefore studied the effects of ribavirin, with or without inhibition of the nonstructural (NS)3/4A protease, on intrahepatic immunity. The intrahepatic immunity of wild-type and NS3/4A-transgenic mice was determined by Western blot, ELISA, flow cytometry, and survival analysis. Various MAVS or TCPTP constructs were injected hydrodynamically to study their relevance. Ribavirin pretreatment was performed in mice expressing a functional or inhibited NS3/4A protease to analyze its effect on NS3/4A-mediated changes. Intrahepatic NS3/4A expression made mice resistant to TNF-α-induced liver damage and caused an alteration of the intrahepatic cytokine (IFN-γ and IL-10) and chemokine (CCL3, CCL17, CCL22, CXCL9, and CXCL11) profiles toward an anti-inflammatory state. Consistent with this, the number of intrahepatic Th1 cells and IFN-γ(+) T cells in NS3/4A-transgenic mice decreased, whereas the amount of Th2 cells increased. These effects could be reversed by injection of uncleavable TCPTP but not uncleavable MAVS and were absent in a mouse expressing a nonfunctional NS3/4A protease. Importantly, the NS3/4A-mediated effects were reversed by ribavirin treatment. Thus, cleavage of TCPTP by NS3/4A induces a shift of the intrahepatic immune response toward a nonantiviral Th2-dominated immunity. These effects are reversed by ribavirin, supporting that ribavirin complements the effects of direct-acting antivirals as an immunomodulatory compound.


Subject(s)
Hepacivirus/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Ribavirin/pharmacology , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antiviral Agents/pharmacology , Cell Differentiation/drug effects , Chemokine CCL17/biosynthesis , Chemokine CCL22/biosynthesis , Chemokine CCL3/biosynthesis , Chemokine CXCL11/biosynthesis , Chemokine CXCL9/biosynthesis , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Th1 Cells , Th2 Cells , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics
8.
J Immunol ; 190(4): 1646-58, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23315077

ABSTRACT

The thymus is a target of multiple pathogens. How the immune system responds to thymic infection is largely unknown. Despite being considered an immune-privileged organ, we detect a mycobacteria-specific T cell response in the thymus following dissemination of Mycobacterium avium or Mycobacterium tuberculosis. This response includes proinflammatory cytokine production by mycobacteria-specific CD4(+) and CD8(+) T cells, which stimulates infected cells and controls bacterial growth in the thymus. Importantly, the responding T cells are mature peripheral T cells that recirculate back to the thymus. The recruitment of these cells is associated with an increased expression of Th1 chemokines and an enrichment of CXCR3(+) mycobacteria-specific T cells in the thymus. Finally, we demonstrate it is the mature T cells that home to the thymus that most efficiently control mycobacterial infection. Although the presence of mature T cells in the thymus has been recognized for some time, to our knowledge, these data are the first to show that T cell recirculation from the periphery to the thymus is a mechanism that allows the immune system to respond to thymic infection. Maintaining a functional thymic environment is essential to maintain T cell differentiation and prevent the emergence of central tolerance to the invading pathogens.


Subject(s)
Cell Movement/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Thymus Gland/immunology , Thymus Gland/microbiology , Animals , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Immunity, Innate , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mycobacterium avium/immunology , Mycobacterium tuberculosis/immunology , Receptors, CXCR3/biosynthesis , T-Lymphocyte Subsets/pathology , Thymus Gland/pathology , Tuberculosis/immunology , Tuberculosis/prevention & control
9.
J Immunol ; 191(8): 4202-10, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24038090

ABSTRACT

During infection with the helminth parasite Schistosoma mansoni, Ab regulates hepatic inflammation, and local production of Ig in the liver appears to play a role in this process. Exploring the development of the B cell response during infection, we found that parasite-specific IgG1-secreting plasma cells appeared first in the hepatic and mesenteric lymph nodes (LNs) and then at later times in the spleen, liver, and bone marrow. The LN B cell population peaked between weeks 10 and 12 of infection, and then contracted at a time that coincided with the expansion of the hepatic IgG1(+) B cell compartment, suggesting that B cells migrate from LNs to liver. CXCL9 and -16 expression in the liver increased during the time frame of B cell recruitment. Expression of the CXCL16 receptor CXCR6 was increased on B cells within the hepatic LNs, but not the mesenteric LNs. CXCR3, the receptor for CXCL9, was broadly expressed on IgG1(+) B cells in LNs and liver during infection. Increased hepatic expression of CXCL9 and -16 failed to occur if the IL-10R was blocked in vivo, an intervention associated with decreased liver B cell infiltration and the development of severe disease. Hepatic LN IgG1(+) cells migrated toward CXCL9 and -16 in vitro and to the liver in a pertussis toxin-sensitive fashion. Our data suggest that the coordinated expression of CXCL9 and -16 in the liver and of CXCR6 and CXCR3 on responding B cells within the hepatic LNs underpins establishment of the hepatic B cell infiltrate during chronic schistosomiasis.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/immunology , Liver/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Adoptive Transfer , Animals , Bone Marrow/immunology , Cell Movement/immunology , Chemokine CXCL16 , Chemokine CXCL6/biosynthesis , Chemokine CXCL9/biosynthesis , Inflammation/immunology , Liver/cytology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pertussis Toxin , Receptors, CXCR/biosynthesis , Receptors, CXCR3/biosynthesis , Receptors, CXCR6 , Receptors, Interleukin-10/biosynthesis , Schistosomiasis mansoni/parasitology , Spleen/cytology , Spleen/immunology
10.
J Immunol ; 188(12): 6247-57, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22586042

ABSTRACT

Recruitment of CD4(+) T cells to infection areas after HSV-2 infection may be one of the mechanisms that account for increased HIV-1 sexual transmission. Lymphocytes recruited by chemokine CXCL9 are known to be important in control of HSV-2 infection in mice, although the underlying mechanism remains to be addressed. Based on our observation that CXCL9 expression is augmented in the cervical mucus of HSV-2-positive women, in this study we demonstrate that HSV-2 infection directly induces CXCL9 expression in primary cervical epithelial cells and cell lines, the principal targets of HSV-2, at both mRNA and protein levels. Further studies reveal that the induction of CXCL9 expression by HSV-2 is dependent upon a binding site for C/EBP-ß within CXCL9 promoter sequence. Furthermore, CXCL9 expression is promoted at the transcriptional level through phosphorylating C/EBP-ß via p38 MAPK pathway, leading to binding of C/EBP-ß to the CXCL9 promoter. Chemotaxis assays indicate that upregulation of CXCL9 expression at the protein level by HSV-2 infection enhances the migration of PBLs and CD4(+) T cells, whereas neutralization of CXCL9 or inhibition of p38-C/EBP-ß pathway can significantly decrease the migration. Our data together demonstrate that HSV-2 induces CXCL9 expression in human cervical epithelial cells by activation of p38-C/EBP-ß pathway through promoting the binding of C/EBP-ß to CXCL9 promoter, which may recruit activated CD4(+) T cells to mucosal HSV-2 infection sites and potentially increase the risk of HIV-1 sexual transmission.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chemokine CXCL9/biosynthesis , Epithelial Cells/virology , Herpes Simplex/metabolism , MAP Kinase Signaling System/immunology , Adult , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/immunology , CD4-Positive T-Lymphocytes/immunology , Cervix Uteri/virology , Chemokine CXCL9/genetics , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Herpes Simplex/immunology , Herpesvirus 2, Human/immunology , Humans , Middle Aged , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Young Adult
11.
Immunopharmacol Immunotoxicol ; 36(6): 412-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25255161

ABSTRACT

Elevation of myeloid-derived suppressor cells (MDSCs) was observed in some viral infectious diseases. In this study, we studied whether ribavirin, a widely used clinical antiviral drug, could impact the differentiation of human MDSCs in vitro. Flow cytometric analysis showed that ribavirin treatment (5-20 µg/ml) significantly enhanced the differentiation of monocytic MDSCs in a dose-dependent manner. The ribavirin-generated MDSCs were immune-suppressive toward autologous T cells. The mRNA expression of some cytokines was further examined by quantitative reverse transcription polymerase chain reaction. We observed a significant down-regulation of chemokine (C-X-C motif) ligand 9 (CXCL9) and CXCL10 mRNA in ribavirin-generated MDSCs, when compared with control. Peripheral blood mononuclear cells from clinical chronic hepatitis C patients subjected to ribavirin therapy also displayed a similar suppression in CXCL9/10 mRNA expression. Administration of recombinant CXCL9/10 proteins clearly counteracted the effect of ribavirin on MDSCs. In summary, this study showed that ribavirin enhanced human MDSCs differentiation in vitro, which may be attribute to the down-regulation of CXCL9/10 expression.


Subject(s)
Antiviral Agents/pharmacology , Cell Differentiation/drug effects , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Myeloid Cells/drug effects , Ribavirin/pharmacology , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Flow Cytometry , Humans , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
12.
Int J Cancer ; 133(11): 2609-18, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23686423

ABSTRACT

Abundant macrophage infiltration in tumors often correlates with a poor prognosis. T cell/histiocyte rich large B cell lymphoma (THRLBCL) is a distinct aggressive B cell lymphoma entity showing a high macrophage content. To further elucidate the role of tumor-associated macrophages in THRLBCL, we performed gene expression profiling of microdissected histiocyte subsets of THRLBCL, nodular lymphocyte predominant Hodgkin lymphoma (NLPHL), Piringer lymphadenitis, sarcoidosis, nonspecific lymphadenitis and monocytes from peripheral blood. In a supervised principal component analysis, histiocytes from THRLBCL were most closely related to epithelioid cells from NLPHL, with both types of cells expressing genes related to proinflammatory and regulatory macrophage activity. Moreover, histiocytes from THRLBCL strongly expressed metal-binding proteins like MT2A, by which histiocytes of THRLBCL can be distinguished from the other histiocyte subsets investigated. Interestingly, the validation at the protein level showed a strong expression of TXN, CXCL9, MT2A and SOD2 not only in macrophages of THRLBCL but also in the tumor cells of NLPHL and classical Hodgkin lymphoma (cHL). Overall, the present findings indicate that macrophages in the microenvironment of THRLBCL have acquired a distinct gene expression pattern that is characterized by a mixed M1/M2 phenotype and a strong expression of several metal binding proteins. The microenvironments in NLPHL and THRLBCL appear to have a similar influence on the macrophage phenotype. The high expression of metal binding proteins in histiocytes of THRLBCL may be diagnostically useful, but a potential pathophysiological role remains to be identified.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Chemokine CXCL9/biosynthesis , Hodgkin Disease/pathology , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Macrophages/metabolism , Metallothionein/biosynthesis , Neoplasm Staging , Prognosis , Superoxide Dismutase/biosynthesis , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thioredoxins/biosynthesis
13.
Biol Blood Marrow Transplant ; 19(3): 378-86, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266741

ABSTRACT

Relapse and graft-versus-host disease remain major problems associated with allogeneic bone marrow (BM) transplantation (allo-BMT) and posttransplantation therapy in patients with multiple myeloma (MM) and other hematologic malignancies. A possible strategy for selectively enhancing the graft-versus-myeloma response and possibly reducing graft-versus-host disease is to increase the migration of alloreactive T cells toward the MM-containing BM. In the present study, we characterized the BM-homing behavior of donor-derived effector T cells in a novel allo-BMT model for the treatment of MM. We observed that posttransplantation immunotherapy consisting of donor lymphocyte infusion (DLI) and vaccination with minor histocompatibility antigen-loaded dendritic cells (DCs) was associated with prolonged survival compared with allo-BMT with no further treatment. Moreover, CD8(+) effector T cells expressing inflammatory homing receptors, including high levels of CD44, LFA-1, and inflammatory chemokine receptors, were recruited to MM-bearing BM. This was paralleled by strongly increased expression of IFN-γ and IFN-γ-inducible chemokines, including CXCL9, CXCL10, and CXCL16, especially in mice treated with DLI plus minor histocompatibility antigen-loaded DC vaccination. Remarkably, expression of the homeostatic chemokine CXCL12 was reduced. Furthermore, IFN-γ and TNF-α induced BM endothelial cells to express high levels of the inflammatory chemokines and reduced or unaltered levels of CXCL12. Finally, presentation of CXCL9 by multiple BM endothelial cell-expressed heparan sulfate proteoglycans triggered transendothelial migration of effector T cells. Taken together, our data demonstrate that both post-transplantation DLI plus miHA-loaded DC vaccination and MM growth result in an increased expression of inflammatory homing receptors on donor T cells, decreased levels of the homeostatic BM-homing chemokine CXCL12, and strong induction of inflammatory chemokines in the BM. Thus, along with increasing the population of alloreactive T cells, post-transplantation immunotherapy also might contribute to a more effective graft-versus-tumor response by switching homeostatic T cell migration to inflammation-driven migration.


Subject(s)
Bone Marrow Transplantation , Bone Marrow/immunology , Dendritic Cells/immunology , Immunotherapy , Multiple Myeloma/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow/pathology , Cell Movement/immunology , Chemokine CXCL10/agonists , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/immunology , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/immunology , Chemokine CXCL16 , Chemokine CXCL6/agonists , Chemokine CXCL6/biosynthesis , Chemokine CXCL6/immunology , Chemokine CXCL9/agonists , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/immunology , Dendritic Cells/chemistry , Dendritic Cells/transplantation , Graft vs Tumor Effect/immunology , Interferon-gamma/agonists , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Transfusion , Mice , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/immunology , Multiple Myeloma/immunology , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Survival Analysis , Transplantation, Homologous , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
14.
Cytokine ; 64(1): 382-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23819906

ABSTRACT

Chemokines have been shown to play an important role in the pathogenesis of pancreatitis, but the role of chemokine CXCL9 in pancreatitis is poorly understood. The aim of this study was to investigate whether CXCL9 was a modulating factor in chronic pancreatitis. Chronic pancreatitis was induced in Sprague-Dawley rats by intraductal infusion of trinitrobenzene sulfonic acid (TNBS) and CXCL9 expression was assessed by immunohistochemistry, Western blot analysis and enzyme linked immunosorbent assay (ELISA). Recombinant human CXCL9 protein (rCXCL9), neutralizing antibody and normal saline (NS) were administered to rats with chronic pancreatitis by subcutaneous injection. The severity of fibrosis was determined by measuring hydroxyproline in pancreatic tissues and histological grading. The effect of rCXCL9 on activated pancreatic stellate cells (PSCs) in vitro was examined and collagen 1α1, TGF-ß1 and CXCR3 expression was assessed by Western blot analysis in isolated rat PSCs. Chronic pancreatic injury in rats was induced after TNBS treatment and CXCL9 protein was markedly upregulated during TNBS-induced chronic pancreatitis. Although parenchymal injury in the pancreas was not obviously affected after rCXCL9 and neutralizing antibody administration, rCXCL9 could attenuate fibrogenesis in TNBS-induced chronic pancreatitis in vivo and exerted antifibrotic effects in vitro, suppressing collagen production in activated PSCs. In conclusion, CXCL9 is involved in the modulation of pancreatic fibrogenesis in TNBS-induced chronic pancreatitis in rats, and may be a therapeutic target in pancreatic fibrosis.


Subject(s)
Chemokine CXCL9/metabolism , Pancreas/metabolism , Pancreatitis, Chronic/metabolism , Animals , Antibodies, Neutralizing/immunology , Cell Proliferation , Chemokine CXCL9/biosynthesis , Collagen Type I/biosynthesis , Fibrosis , Hydroxyproline/analysis , Male , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/immunology , Rats , Rats, Sprague-Dawley , Receptors, CXCR3/biosynthesis , Transforming Growth Factor beta1/biosynthesis , Trinitrobenzenesulfonic Acid
15.
J Immunol ; 185(3): 1864-77, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20585034

ABSTRACT

The IFN-gamma-inducible chemokines CXCL9 and CXCL10 are implicated in the pathogenesis of T cell-mediated immunity in the CNS. However, in various CNS immune pathologies the cellular localization of these chemokines differs, with CXCL9 produced by macrophage/microglia whereas CXCL10 is produced by both macrophage/microglia and astrocytes. In this study, we determined the mechanism for the microglial cell-restricted expression of the Cxcl9 gene induced by IFN-gamma. In cultured glial cells, the induction of the CXCL9 (in microglia) and CXCL10 (in microglia and astrocytes) mRNAs by IFN-gamma was not inhibited by cycloheximide. Of various transcription factors involved with IFN-gamma-mediated gene regulation, PU.1 was identified as a constitutively expressed NF in microglia but not in astrocytes. STAT1 and PU.1 bound constitutively to the Cxcl9 gene promoter in microglia, and this increased significantly following IFN-gamma treatment with IFN regulatory factor-8 identified as an additional late binding factor. However, in astrocytes, STAT1 alone bound to the Cxcl9 gene promoter. STAT1 was critical for IFN-gamma induction of both the Cxcl9 and Cxcl10 genes in microglia and in microglia and astrocytes, respectively. The small interfering RNA-mediated knockdown of PU.1 in microglia markedly impaired IFN-gamma-induced CXCL9 but not STAT1 or IFN regulatory factor-8. Cells of the D1A astrocyte line showed partial reprogramming to a myeloid-like phenotype posttransduction with PU.1 and, in addition to the expression of CD11b, acquired the ability to produce CXCL9 in response to IFN-gamma. Thus, PU.1 not only is crucial for the induction of CXCL9 by IFN-gamma in microglia but also is a key determinant factor for the cell-specific expression of this chemokine by these myeloid cells.


Subject(s)
Central Nervous System/immunology , Central Nervous System/metabolism , Chemokine CXCL9/biosynthesis , Interferon-gamma/physiology , Microglia/immunology , Microglia/metabolism , Myeloid Cells/immunology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Cell Line , Cell Lineage/immunology , Cells, Cultured , Central Nervous System/cytology , Chemokine CXCL10/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , STAT1 Transcription Factor/physiology
16.
Clin Dev Immunol ; 2012: 934161, 2012.
Article in English | MEDLINE | ID: mdl-22969819

ABSTRACT

Abnormalities in monocytes and in peripheral blood dendritic cells (DC) subsets have been reported in systemic lupus erythematosus (SLE). We aim to clarify the tolerogenic or inflammatory role of these cells based on ICOSL or IFN-α and chemokine mRNA expression, respectively, after cell purification. The study included 18 SLE patients with active disease (ASLE), 25 with inactive disease (ISLE), and 30 healthy controls (HG). In purified plasmacytoid DC (pDC) was observed a lower ICOSL mRNA expression in ASLE and an increase in ISLE; similarly, a lower ICOSL mRNA expression in monocytes of ALSE patients was found. However, a higher ICOSL mRNA expression was observed in ASLE compared to HG in myeloid DCs. Interestingly, clinical parameters seem to be related with ICOSL mRNA expression. Regarding the inflammatory activity it was observed in purified monocytes and CD14(-/low) CD16(+) DCs an increase of CCL2, CXCL9, and CXCL10 mRNA expression in ASLE compared to HG. In myeloid DC no differences were observed regarding chemokines, and IFN-α mRNA expression. In pDC, a higher IFN-α mRNA expression was observed in ASLE. Deviations in ICOSL, chemokine, and IFN-α mRNA expression in peripheral blood monocytes and dendritic cells subpopulations in SLE appear to be related to disease activity.


Subject(s)
Dendritic Cells/immunology , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/immunology , Adult , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Female , Humans , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/genetics , Inflammation/immunology , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/blood , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Self Tolerance/immunology
17.
Proc Natl Acad Sci U S A ; 106(46): 19455-60, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19892741

ABSTRACT

Treatment of mice bearing orthotopic, metastatic tumors with anti-CD40 antibody resulted in only partial, transient anti-tumor effects whereas combined treatment with IL-2/anti-CD40, induced tumor regression. The mechanisms for these divergent anti-tumor responses were examined by profiling tumor-infiltrating leukocyte subsets and chemokine expression within the tumor microenvironment after immunotherapy. IL-2/anti-CD40, but not anti-CD40 alone, induced significant infiltration of established tumors by NK and CD8(+) T cells. To further define the role of chemokines in leukocyte recruitment into tumors, we evaluated anti-tumor responses in mice lacking the chemokine receptor, CCR2. The anti-tumor effects and leukocyte recruitment mediated by anti-CD40 alone, were completely abolished in CCR2(-/-) mice. In contrast, IL-2/anti-CD40-mediated leukocyte recruitment and reductions in primary tumors and metastases were maintained in CCR2(-/-) mice. Treatment of mice with IL-2/anti-CD40, but not anti-CD40 alone, also caused an IFN-gamma-dependent increase in the expression of multiple Th1 chemokines within the tumor microenvironment. Interestingly, although IL-2/anti-CD40 treatment increased Tregs in the spleen, it also caused a coincident IFN-gamma-dependent reduction in CD4(+)/FoxP3(+) Tregs, myeloid-derived suppressor cells and Th2 chemokine expression specifically within the tumor microenvironment that was not observed after treatment with anti-CD40 alone. Similar effects were observed using IL-15 in combination with anti-CD40. Taken together, our data demonstrate that IL-2/anti-CD40, but not anti-CD40 alone, can preferentially reduce the overall immunosuppressive milieu within the tumor microenvironment. These results suggest that the use of anti-CD40 in combination with IL-2 or IL-15 may hold substantially more promise for clinical cancer treatment than anti-CD40 alone.


Subject(s)
Antibodies/therapeutic use , CD40 Antigens/agonists , Immunosuppression Therapy/methods , Interleukin-2/therapeutic use , Neoplasms/therapy , Animals , Arginase/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL5/biosynthesis , Chemokine CXCL9/biosynthesis , Chemokines/biosynthesis , Chemokines, CC/biosynthesis , Drug Synergism , Lymphocytes, Tumor-Infiltrating/immunology , Macrophage Inflammatory Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Receptors, CCR2/biosynthesis , Receptors, CCR2/genetics , Receptors, Cytokine/biosynthesis
18.
Hepatogastroenterology ; 59(116): 1245-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22580676

ABSTRACT

BACKGROUND/AIMS: The aim of this study was to investigate the effect of HBV-induced Mig and role of autophagy in the process. METHODOLOGY: Adxsi-1.3 x HBV plasmid was constructed and identified. The three cell lines (L02, HepG2, SMMC-7721) were infected with adenovirus-HBV. HBsAg and HBeAg were assessed by electrochemiluminescence immunoassay. HBV DNA, HBx, Beclin 1 and Mig expression were detected by quantitative real-Time PCR, western blotting and ELISA. The level of autophagy was evaluated by transmission electron microscope. RESULTS: Human fetal liver cells and hepatocellular carcinoma cells were successfully transfected with overlength HBV genome using an adenovirus vector (Ad-HBV). Ad-HBV induced Mig production and cell autophagy through up-regulation of Beclin 1 expression. We further demonstrated that the increased autophagy extent was in association with HBV-induced Mig expression. CONCLUSIONS: Autophagy may be a crucial intracellular mechanism of Mig induction in response to HBV infection. The results provide new insights into the pathogenesis of HBV.


Subject(s)
Adenoviridae/genetics , Autophagy/physiology , Chemokine CXCL9/biosynthesis , Hepatitis B virus/pathogenicity , Apoptosis Regulatory Proteins/biosynthesis , Beclin-1 , Cells, Cultured , Class III Phosphatidylinositol 3-Kinases/physiology , DNA, Viral/analysis , Genetic Vectors , Hep G2 Cells , Hepatitis B Surface Antigens/analysis , Hepatitis B e Antigens/analysis , Humans , Membrane Proteins/biosynthesis , Trans-Activators/analysis , Transfection , Viral Regulatory and Accessory Proteins
19.
Innate Immun ; 28(5): 155-163, 2022 07.
Article in English | MEDLINE | ID: mdl-35548957

ABSTRACT

Th17 cells represent important immune cells. Ursolic acid (UA) can regulate immune cell activities. This study was aimed to explore the effects of UA on Th17 cell differentiation and Schwann cell(SCs)-mediated migration and the potential mechanism. Naïve CD4+ T cells were isolated from rat peripheral blood, induced for Th17 cell differentiation, and treated with UA. The proportion of Th17 cells was detected by flow cytometry assay. SCs were co-cultured with Th17 cells. Th17 cell migration was detected by Transwell assay. The molecule expression was determined by Western blot and qRT-PCR. UA inhibited the Th17 cell differentiation and suppressed the STAT3/RORγt pathway. STAT3 overexpression up-regulated p-STAT3 and RORγt expression and promoted Th17 cell differentiation under the UA treatment. In LPS- and IFN-γ-stimulated-SCs, UA suppressed the expression of chemokines CXCL9/10, but had no significant effect of CCL20 expression. The expression CXCL9/10 receptor CXCR3 was higher in Th17 cells than that in Treg cells, while the expression CCL20 receptor CCR6 was lower in Th17 cells than that in Treg cells. UA, anti-CXCR3, and anti-CCR6 treatment inhibited SCs-mediated Th17 cell migration, and anti-CXCR3 exerted stronger inhibitory effect than Anti-CCR6. UA inhibited Th17 cell differentiation through STAT3/RORγt pathway and suppressed Th17 cell migration through down-regulating CXCL9/10 expression in SCs.


Subject(s)
Chemokine CXCL10 , Chemokine CXCL9 , Nuclear Receptor Subfamily 1, Group F, Member 3 , STAT3 Transcription Factor , Schwann Cells , Th17 Cells , Triterpenes , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/metabolism , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Rats , STAT3 Transcription Factor/metabolism , Schwann Cells/cytology , Schwann Cells/drug effects , Schwann Cells/metabolism , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/metabolism , Triterpenes/pharmacology , Ursolic Acid
20.
Mol Pharmacol ; 79(3): 586-95, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173040

ABSTRACT

Herein we provide evidence for the coexpression of two distinct prostacyclin (PGI(2)) receptors (IP) on BEAS-2B human airway epithelial cells. IP receptor heterogeneity initially was suggested by the finding that the rank orders of potency of PGI(2) and three structurally similar analogs [taprostene, iloprost, 15-deoxy-16-(m-tolyl)-17,18,19,20-tetranorisocarbacyclin (15-deoxy-TIC)] for the inhibition of chemokine (CXCL9 and CXCL10) release and for transcriptional activation/augmentation of cAMP response element and glucocorticoid response element luciferase reporters were distinct. Indeed, PGI(2), taprostene, and iloprost activated both reporters whereas 15-deoxy-TIC was inert. Conversely, 15-deoxy-TIC, PGI(2), and taprostene (but not iloprost) suppressed chemokine release. Further experiments established that iloprost did not antagonize the inhibitory effect taprostene or 15-deoxy-TIC on chemokine output. Likewise, 15-deoxy-TIC failed to antagonize taprostene- and iloprost-induced reporter transactivation. Thus, iloprost- and 15-deoxy-TIC-induced responses were apparently mediated via pharmacologically distinct receptors. In human embryonic kidney 293 cells overexpressing the human recombinant IP receptor receptor, 15-deoxy-TIC was considerably less potent (>10,000-fold) than iloprost and taprostene in promoting cAMP accumulation, yet in BEAS-2B cells, these analogs were equipotent. IP receptor heterogeneity was also supported by the finding that the affinity of the IP receptor antagonist R-3-(4-fluorophenyl)-2-[5-(4-fluorophenyl)-benzofuran-2-yl-methoxycarbonyl-amino] propionic acid (RO3244794) for the receptor mediating inhibition of chemokine release was approximately 10-fold lower than for the receptor mediating both transcriptional outputs. Finally, small interfering RNAs directed against the IP receptor gene, PTGIR, failed to block the suppression of chemokine output induced by taprostene and 15-deoxy-TIC, whereas taprostene- and iloprost-induced transcriptional responses were markedly attenuated. Collectively, these results indicate that PGI(2), taprostene and 15-deoxy-TIC suppress chemokine release from BEAS-2B cells by interacting with a novel IP receptor that we denote here as the "IP(2)" subtype.


Subject(s)
Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Receptors, Prostaglandin/metabolism , Respiratory Mucosa/metabolism , Benzofurans/pharmacology , Cells, Cultured , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/antagonists & inhibitors , Chemokine CXCL9/biosynthesis , Cyclic AMP/metabolism , Cyclic AMP/physiology , Dose-Response Relationship, Drug , Epoprostenol/analogs & derivatives , Epoprostenol/metabolism , Epoprostenol/pharmacology , HEK293 Cells , Humans , Iloprost/pharmacology , Propionates/pharmacology , Receptors, Epoprostenol , Receptors, Prostaglandin/drug effects , Receptors, Prostaglandin/physiology , Recombinant Proteins/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/physiology
SELECTION OF CITATIONS
SEARCH DETAIL