Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(3): e0120917, 2015.
Article in English | MEDLINE | ID: mdl-25807374

ABSTRACT

Synovial fibroblasts in persistent inflammatory arthritis have been suggested to have parallels with cancer growth and wound healing, both of which involve a stereotypical serum response programme. We tested the hypothesis that a serum response programme can be used to classify diseased tissues, and investigated the serum response programme in fibroblasts from multiple anatomical sites and two diseases. To test our hypothesis we utilized a bioinformatics approach to explore a publicly available microarray dataset including rheumatoid arthritis (RA), osteoarthritis (OA) and normal synovial tissue, then extended those findings in a new microarray dataset representing matched synovial, bone marrow and skin fibroblasts cultured from RA and OA patients undergoing arthroplasty. The classical fibroblast serum response programme discretely classified RA, OA and normal synovial tissues. Analysis of low and high serum treated fibroblast microarray data revealed a hierarchy of control, with anatomical site the most powerful classifier followed by response to serum and then disease. In contrast to skin and bone marrow fibroblasts, exposure of synovial fibroblasts to serum led to convergence of RA and OA expression profiles. Pathway analysis revealed three inter-linked gene networks characterising OA synovial fibroblasts: Cell remodelling through insulin-like growth factors, differentiation and angiogenesis through _3 integrin, and regulation of apoptosis through CD44. We have demonstrated that Fibroblast serum response signatures define disease at the tissue level, and that an OA specific, serum dependent repression of genes involved in cell adhesion, extracellular matrix remodelling and apoptosis is a critical discriminator between cultured OA and RA synovial fibroblasts.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Osteoarthritis/pathology , Actin Cytoskeleton/metabolism , Arthritis, Rheumatoid/metabolism , Bone Marrow Cells/cytology , Cells, Cultured , Databases, Factual , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Integrin beta3/pharmacology , Oligonucleotide Array Sequence Analysis , Osteoarthritis/metabolism , Principal Component Analysis , Signal Transduction/drug effects , Skin/cytology , Somatomedins/pharmacology , Synovial Membrane/cytology , Transcriptome
2.
Cancer Immunol Res ; 3(10): 1138-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25711537

ABSTRACT

Infusing virus-specific T cells is effective treatment for rare Epstein-Barr virus (EBV)-associated posttransplant lymphomas, and more limited success has been reported using this approach to treat a far more common EBV-associated malignancy, nasopharyngeal carcinoma (NPC). However, current approaches using EBV-transformed lymphoblastoid cell lines to reactivate EBV-specific T cells for infusion take 2 to 3 months of in vitro culture and favor outgrowth of T cells targeting viral antigens expressed within EBV(+) lymphomas, but not in NPC. Here, we explore T-cell receptor (TCR) gene transfer to rapidly and reliably generate T cells specific for the NPC-associated viral protein LMP2. We cloned a human leukocyte antigen (HLA) A*1101-restricted TCR, which would be widely applicable because 40% of NPC patients carry this HLA allele. Studying both the wild-type and modified forms, we have optimized expression of the TCR and demonstrated high-avidity antigen-specific function (proliferation, cytotoxicity, and cytokine release) in both CD8(+) and CD4(+) T cells. The engineered T cells also inhibited LMP2(+) epithelial tumor growth in a mouse model. Furthermore, transduced T cells from patients with advanced NPC lysed LMP2-expressing NPC cell lines. Using this approach, within a few days large numbers of high-avidity LMP2-specific T cells can be generated reliably to treat NPC, thus providing an ideal clinical setting to test TCR gene transfer without the risk of autoimmunity through targeting self-antigens.


Subject(s)
Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , HLA-A Antigens/immunology , Herpesvirus 4, Human/immunology , Nasopharyngeal Neoplasms/etiology , Receptors, Antigen, T-Cell/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression , Gene Transfer Techniques , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Mice , Molecular Sequence Data , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/therapy , Transduction, Genetic , Tumor Burden , Viral Matrix Proteins/immunology , Xenograft Model Antitumor Assays
3.
Am J Pathol ; 180(3): 1215-1222, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22226739

ABSTRACT

The substantial T lymphocyte infiltrate found in cases of nasopharyngeal carcinoma (NPC) has been implicated in the promotion of both tumor growth and immune escape. Conversely, because malignant NPC cells harbor the Epstein-Barr virus, this tumor is a candidate for virus-specific T cell-based therapies. Preventing the accumulation of tumor-promoting T cells or enhancing the recruitment of tumor-specific cytotoxic T cells offers therapeutic potential. However, the mechanisms involved in T cell recruitment to this tumor are poorly understood. Comparing memory T cell subsets that have naturally infiltrated NPC tissue with their counterparts from matched blood revealed enrichment of CD8(+), CD4(+), and regulatory T cells expressing the chemokine receptor CXCR6 in tumor tissue. CD8(+) and (nonregulatory) CD4(+) T cells also were more frequently CCR5(+) in tumor than in blood. Ex vivo studies demonstrated that both receptors were functional. CXCL16 and CCL4, unique chemokine ligands for CXCR6 and CCR5, respectively, were expressed by the malignant cells in tumor tissue from the majority of NPC cases, as was another CCR5 ligand, CCL5. The strongest expression of CXCL16 was found on tumor-infiltrating cells. CCL4 was detected on the tumor vasculature in a majority of cases. These findings suggest that CXCR6 and CCR5 play important roles in T cell recruitment and/or retention in NPC and have implications for the pathogenesis and treatment of this tumor.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Nasopharyngeal Neoplasms/immunology , Receptors, CCR5/metabolism , Receptors, Chemokine/metabolism , Receptors, Virus/metabolism , T-Lymphocyte Subsets/immunology , Carcinoma , Cell Line, Tumor , Humans , Immunologic Memory/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Nasopharyngeal Carcinoma , Receptors, CCR5/physiology , Receptors, CXCR6 , Receptors, Chemokine/physiology , Receptors, Virus/physiology
4.
Eur Urol ; 61(2): 385-94, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079021

ABSTRACT

BACKGROUND: Evidence suggests that some patients with renal cell carcinoma (RCC) respond to immunomodulatory therapies that activate T lymphocytes. A prerequisite for effective T cell therapy is efficient targeting of effector T cells to the tumour site, yet the molecular basis of T cell recruitment to RCC is unknown. Furthermore, some T cells that naturally infiltrate this cancer are regulatory T cells (Tregs) that may suppress antitumour immune responses. OBJECTIVE: Determine the mechanisms of effector and regulatory T cell recruitment to RCC to allow targeted therapy that promotes local anti-tumour immunity. DESIGN, SETTING, AND PARTICIPANTS: Tumour-infiltrating and peripheral blood T cells were collected from 70 patients undergoing nephrectomy for RCC. MEASUREMENTS: T cells were analysed by multicolour flow cytometry for expression of 19 chemokine receptors and 7 adhesion molecules. Receptors that were expressed at higher levels on tumour-infiltrating lymphocytes (TILs) compared with matched peripheral blood lymphocytes (PBLs) were analysed further for their ability to mediate migration responses in TILs and for expression of corresponding ligands in tumour tissue. RESULTS AND LIMITATIONS: Three chemokine receptors-CCR5, CXCR3, and CXCR6-were significantly overexpressed on TILs compared with matched PBLs (n=16 cases) and were capable of promoting migration in vitro. Their corresponding ligands CCL4-5, CXCL9-11, and CXCL16 were all detected in RCC tissue. However, since they were present in all cases studied, it was not possible to correlate ligand expression with levels of T cell infiltration. Foxp3(+) Tregs were enriched within TILs compared with matched PBLs and expressed high levels of CCR5, CXCR3, and CXCR6, as well as CCR6, the ligand for which (CCL20) was detectable in RCC tissue. CONCLUSIONS: Our data support a role for CCR5, CXCR3, and CXCR6 in the selective recruitment of T cells into RCC tissue and, together with CCR6, in the recruitment of Tregs.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, CCR5/immunology , Receptors, CCR6/immunology , Receptors, CXCR3/immunology , Receptors, Chemokine/immunology , Receptors, Virus/immunology , Adult , Aged , Aged, 80 and over , Cell Movement/immunology , Female , Flow Cytometry/methods , Forkhead Transcription Factors/immunology , Humans , Male , Middle Aged , Receptors, CXCR6 , T-Lymphocytes/immunology
5.
Arthritis Rheum ; 60(6): 1604-14, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19479862

ABSTRACT

OBJECTIVE: High expression of galectin 3 at sites of joint destruction in rheumatoid arthritis (RA) suggests that galectin 3 plays a role in RA pathogenesis. Previous studies have demonstrated the effects of galectins on immune cells, such as lymphocytes and macrophages. This study was undertaken to investigate the hypothesis that galectin 3 induces proinflammatory effects in RA by modulating the pattern of cytokine and chemokine production in synovial fibroblasts. METHODS: Matched samples of RA synovial and skin fibroblasts were pretreated with galectin 3 or tumor necrosis factor alpha (TNFalpha), and the levels of a panel of cytokines, chemokines, and matrix metalloproteinases (MMPs) were determined using enzyme-linked immunosorbent assays and multiplex assays. Specific inhibitors were used to dissect signaling pathways, which were confirmed by Western blotting and NF-kappaB activation assay. RESULTS: Galectin 3 induced secretion of interleukin-6 (IL-6), granulocyte-macrophage colony-stimulating factor, CXCL8, and MMP-3 in both synovial and skin fibroblasts. By contrast, galectin 3-induced secretion of TNFalpha, CCL2, CCL3, and CCL5 was significantly greater in synovial fibroblasts than in skin fibroblasts. TNFalpha blockade ruled out autocrine TNFalpha-stimulated induction of chemokines. The MAPKs p38, JNK, and ERK were necessary for IL-6 production, but phosphatidylinositol 3-kinase (PI 3-kinase) was required for selective CCL5 induction. NF-kappaB activation was required for production of both IL-6 and CCL5. CONCLUSION: Our findings indicate that galectin 3 promotes proinflammatory cytokine secretion by tissue fibroblasts. However, galectin 3 induces the production of mononuclear cell-recruiting chemokines uniquely from synovial fibroblasts, but not matched skin fibroblasts, via a PI 3-kinase signaling pathway. These data provide further evidence of the role of synovial fibroblasts in regulating the pattern and persistence of the inflammatory infiltrate in RA and suggest a new and important functional consequence of the observed high expression of galectin 3 in the rheumatoid synovium.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokines/metabolism , Cytokines/metabolism , Fibroblasts/metabolism , Galectin 3/physiology , Signal Transduction/physiology , Synovial Membrane/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Chemokine CCL5/metabolism , Fibroblasts/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Skin/cytology , Skin/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Arthritis Res Ther ; 10(2): R47, 2008.
Article in English | MEDLINE | ID: mdl-18433499

ABSTRACT

INTRODUCTION: A surprising feature of the inflammatory infiltrate in rheumatoid arthritis is the accumulation of neutrophils within synovial fluid and at the pannus cartilage boundary. Recent findings suggest that a distinct subset of IL-17-secreting T-helper cells (TH17 cells) plays a key role in connecting the adaptive and innate arms of the immune response and in regulating neutrophil homeostasis. We therefore tested the hypothesis that synovial fibroblasts bridge the biological responses that connect TH17 cells to neutrophils by producing neutrophil survival factors following their activation with IL-17. METHODS: IL-17-expressing cells in the rheumatoid synovium, and IL-17-expressing cells in the peripheral blood, and synovial fluid were examined by confocal microscopy and flow cytometry, respectively. Peripheral blood neutrophils were cocultured either with rheumatoid arthritis synovial fibroblasts (RASF) or with conditioned medium from RASF that had been pre-exposed to recombinant human IL-17, TNFalpha or a combination of the two cytokines. Neutrophils were harvested and stained with the vital mitochondrial dye 3,3'-dihexyloxacarbocyanine iodide before being enumerated by flow cytometry. RESULTS: TH17-expressing CD4+ cells were found to accumulate within rheumatoid synovial tissue and in rheumatoid arthritis synovial fluid. RASF treated with IL-17 and TNFalpha (RASFIL-17/TNF) effectively doubled the functional lifespan of neutrophils in coculture. This was entirely due to soluble factors secreted from the fibroblasts. Specific depletion of granulocyte-macrophage colony-stimulating factor from RASFIL-17/TNF-conditioned medium demonstrated that this cytokine accounted for approximately one-half of the neutrophil survival activity. Inhibition of phosphatidylinositol-3-kinase and NF-kappaB pathways showed a requirement for both signalling pathways in RASFIL-17/TNF-mediated neutrophil rescue. CONCLUSION: The increased number of neutrophils with an extended lifespan found in the rheumatoid synovial microenvironment is partly accounted for by IL-17 and TNFalpha activation of synovial fibroblasts. TH17-expressing T cells within the rheumatoid synovium are likely to contribute significantly to this effect.


Subject(s)
Arthritis, Rheumatoid/immunology , Fibroblasts/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Arthritis, Rheumatoid/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Survival/physiology , Female , Flow Cytometry , Humans , Male , Microscopy, Confocal , Middle Aged , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , T-Lymphocyte Subsets/metabolism
7.
Arthritis Rheum ; 58(3): 707-17, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18311795

ABSTRACT

OBJECTIVE: Fibroblast-like synoviocytes (FLS) are potentially directly involved in the propagation of inflammation. We have previously shown evidence of an expanded activated population of natural killer (NK) cells in spondylarthritis (SpA) patients. In the present study, we sought to determine whether the interaction between NK cells and FLS from SpA patients results in a proinflammatory response. METHODS: Autologous NK cells and FLS were obtained from 6 patients with SpA, 4 patients with rheumatoid arthritis (RA), and 8 patients with osteoarthritis (OA). Physical interactions between NK cells and FLS were studied by time-lapse phase-contrast microscopy. Fluorescence-activated cell sorting was used to study the activation, proliferation, and survival of NK cells in contact with FLS. Cytokine and stromal factor production were measured by a multiple cytokine bead assay. RESULTS: NK cells both adhered to and migrated beneath the FLS monolayer (pseudoemperipolesis). FLS from SpA and RA patients supported increased pseudoemperipolesis, activation, cytokine production, and survival of NK cells. The production of proinflammatory cytokines, including interleukin-6 (IL-6), IL-8, IL-1beta, and IL-15, was increased in cocultures of NK cells and FLS, particularly in those from RA and SpA patients. Production of interferon-gamma, RANTES, and matrix metalloproteinase 3 (MMP-3) by NK cell and FLS coculture was greatest in SpA patients. Surface expression of IL-15 on FLS was significantly increased in SpA and RA patients, but not OA patients. Blockade with an IL-15 monoclonal antibody resulted in increased apoptosis of NK cells. CONCLUSION: FLS promote the migration, activation, and survival of NK cells. The interaction of NK cells with FLS results in increased IL-15 expression by FLS and the production of proinflammatory chemokines, cytokines, and MMPs, which may contribute to joint inflammation. This response was much more marked in SpA and RA patients as compared with OA patients.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cytokines/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Spondylarthritis/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Adult , Aged , Arthritis, Rheumatoid/pathology , Cell Communication/physiology , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-15/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Matrix Metalloproteinase 3/metabolism , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Spondylarthritis/pathology
8.
Arthritis Res Ther ; 8(4): R108, 2006.
Article in English | MEDLINE | ID: mdl-16846535

ABSTRACT

Stromal cells such as fibroblasts play an important role in defining tissue-specific responses during the resolution of inflammation. We hypothesized that this involves tissue-specific regulation of glucocorticoids, mediated via differential regulation of the enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). Expression, activity and function of 11beta-HSD1 was assessed in matched fibroblasts derived from various tissues (synovium, bone marrow and skin) obtained from patients with rheumatoid arthritis or osteoarthritis. 11beta-HSD1 was expressed in fibroblasts from all tissues but mRNA levels and enzyme activity were higher in synovial fibroblasts (2-fold and 13-fold higher mRNA levels in dermal and synovial fibroblasts, respectively, relative to bone marrow). Expression and activity of the enzyme increased in all fibroblasts following treatment with tumour necrosis factor-alpha or IL-1beta (bone marrow: 8-fold and 37-fold, respectively, compared to vehicle; dermal fibroblasts: 4-fold and 14-fold; synovial fibroblasts: 7-fold and 31-fold; all P < 0.01 compared with vehicle). Treatment with IL-4 or interferon-gamma was without effect, and there was no difference in 11beta-HSD1 expression between fibroblasts (from any site) obtained from patients with rheumatoid arthritis or osteoarthritis. In the presence of 100 nmol/l cortisone, IL-6 production--a characteristic feature of synovial derived fibroblasts--was significantly reduced in synovial but not dermal or bone marrow fibroblasts. This was prevented by co-treatment with an 11beta-HSD inhibitor, emphasizing the potential for autocrine activation of glucocorticoids in synovial fibroblasts. These data indicate that differences in fibroblast-derived glucocorticoid production (via the enzyme 11beta-HSD1) between cells from distinct anatomical locations may play a key role in the predeliction of certain tissues to develop persistent inflammation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Arthritis, Rheumatoid/enzymology , Fibroblasts/enzymology , Osteoarthritis/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Arthritis, Rheumatoid/metabolism , Bone Marrow/enzymology , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Cortisone/pharmacology , Glucocorticoids/biosynthesis , Humans , Interleukin-1beta/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Osteoarthritis/metabolism , RNA, Messenger/metabolism , Skin/enzymology , Skin/metabolism , Skin/pathology , Synovial Membrane/enzymology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology
9.
Arthritis Rheum ; 54(7): 2096-108, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16802344

ABSTRACT

OBJECTIVE: Synovial fibroblasts share a number of phenotype markers with fibroblasts derived from bone marrow. In this study we investigated the role of matched fibroblasts obtained from 3 different sources (bone marrow, synovium, and skin) to test the hypothesis that synovial fibroblasts share similarities with bone marrow-derived fibroblasts in terms of their ability to support survival of T cells and neutrophils. METHODS: Matched synovial, bone marrow, and skin fibroblasts were established from 8 different patients with rheumatoid arthritis who were undergoing knee or hip surgery. Resting or activated fibroblasts were cocultured with either CD4 T cells or neutrophils, and the degree of leukocyte survival, apoptosis, and proliferation were measured. RESULTS: Fibroblasts derived from all 3 sites supported increased survival of CD4 T cells, mediated principally by interferon-beta. However, synovial and bone marrow fibroblasts shared an enhanced site-specific ability to maintain CD4 T cell survival in the absence of proliferation, an effect that was independent of fibroblast activation or proliferation but required direct T cell-fibroblast cell contact. In contrast, fibroblast-mediated neutrophil survival was less efficient, being independent of the site of origin of the fibroblast but dependent on prior fibroblast activation, and mediated solely by soluble factors, principally granulocyte-macrophage colony-stimulating factor. CONCLUSION: These results suggest an important functional role for fibroblasts in the differential accumulation of leukocyte subsets in a variety of tissue microenvironments. The findings also provide a potential explanation for site-specific differences in the pattern of T cell and neutrophil accumulation observed in chronic inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid/pathology , Bone Marrow/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Communication/physiology , Fibroblasts/physiology , Neutrophils/pathology , Synovial Membrane/cytology , Apoptosis/physiology , Arthritis, Rheumatoid/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cell Survival/physiology , Coculture Techniques , Cytokines/pharmacology , Fibroblasts/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Interferon-beta/physiology , Neutrophils/drug effects , Neutrophils/metabolism , Skin/metabolism , Skin/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
10.
Trends Immunol ; 26(3): 150-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15745857

ABSTRACT

To navigate into and within tissues, leukocytes require guidance cues that enable them to recognize which tissues to enter and which to avoid. Such cues are partly provided at the time of extravasation from blood by an endothelial address code on the luminal surface of the vascular endothelium. Here, we review the evidence that fibroblasts help define an additional stromal address code that directs leukocyte behaviour within tissues. We examine how this stromal code regulates site-specific leukocyte accumulation, differentiation and survival in a variety of physiological stromal niches, and how the aberrant expression of components of this code in the wrong tissue at the wrong time contributes to the persistence of chronic inflammatory diseases.


Subject(s)
Fibroblasts/immunology , Stromal Cells/immunology , Dendritic Cells/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiology , Fibroblasts/physiology , Leukocytes/immunology , Leukocytes/physiology , Models, Biological , Stromal Cells/physiology , T-Lymphocytes/immunology
11.
Joint Bone Spine ; 72(1): 10-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15681242

ABSTRACT

Rheumatoid arthritis (RA) is a debilitating, chronic, persistent inflammatory disease that is characterised by painful and swollen joints. The aetiology of RA is unknown, however whereas past research has concentrated on the role of immune or inflammatory infiltrating cells in inflammation, it is becoming clear that stromal cells play a critical part in regulating the quality and duration of an inflammatory response. In this review we assess the role of fibroblasts within the inflamed synovium in modulating immune responses; in particular we examine the role of stromal cells in the switch from resolving to persistent inflammation as is found in the rheumatoid synovium.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cell Communication/immunology , Leukocytes/cytology , Stromal Cells/cytology , Animals , Chronic Disease , Humans , Leukocytes/immunology , Synovitis/immunology , Synovitis/pathology
12.
Arthritis Rheum ; 48(9): 2472-82, 2003 Sep.
Article in English | MEDLINE | ID: mdl-13130466

ABSTRACT

OBJECTIVE: A characteristic feature of the inflammatory infiltrate in rheumatoid arthritis is the segregation of CD4 and CD8 T lymphocyte subsets into distinct microdomains within the inflamed synovium. The aim of this study was to test the hypothesis that chemokines in general and stromal cell-derived factor 1 (SDF-1; CXCL12) in particular are responsible for generating this distinctive microcompartmentalization. METHODS: We examined how synovial CD4/CD8 T cell subsets interacted in coculture assays with fibroblasts derived from chronic inflammatory synovial lesions and normal synovial tissue as well as from fetal lung and adult skin. We used the ability of T cells to migrate beneath fibroblasts (a process called pseudoemperipolesis) as an in vitro marker of T cell accumulation within synovial tissue. RESULTS: Rheumatoid fibroblast-like synoviocytes (FLS) displayed a unique ability to support high levels of CD4 and CD8 T cell pseudoemperipolesis. Nonrheumatoid FLS as well as fetal lung fibroblasts supported low levels of pseudoemperipolesis, while skin-derived fibroblasts were unable to do so. CD8 T cells migrated under fibroblasts more efficiently and at a higher velocity than CD4 T cells, a feature that was intrinsic to CD8 T cells. Rheumatoid fibroblasts constitutively produced high levels of SDF-1 (CXCL12), which was functionally important, since blocking studies showed reductions in T cell pseudoemperipolesis to levels seen in nonrheumatoid FLS. Rheumatoid fibroblasts also constitutively produced high levels of vascular cell adhesion molecule 1 (VCAM-1; CD106), but this did not contribute to T cell pseudoemperipolesis, unlike the case for B cells, which require SDF-1 (CXCL12)-CXCR4 and CD49d-VCAM-1 (CD106) interactions. Importantly, only combinations of rheumatoid FLS and rheumatoid-derived synovial fluid T cells supported pseudoemperipolesis when examined ex vivo, confirming the in vivo relevance of these findings. CONCLUSION: These studies demonstrate that features intrinsic to both fibroblasts (the production of SDF-1) and CD8/CD4 T cells (the expression of CXCR4) are responsible for the characteristic pattern of T lymphocyte accumulation seen in the rheumatoid synovium. These findings suggest that the SDF-1/CXCR4 ligand/receptor pair is likely to play an important functional role in T lymphocyte accumulation and positioning within the rheumatoid synovium.


Subject(s)
Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Movement/immunology , Chemokines, CXC/metabolism , Synovial Membrane/immunology , Arthritis, Rheumatoid/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Chemokine CXCL12 , Chemokines, CXC/genetics , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression/immunology , Humans , Pertussis Toxin/pharmacology , Receptors, CXCR4/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , Synovial Membrane/cytology , Synovial Membrane/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
13.
Thromb Haemost ; 90(4): 688-97, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14515190

ABSTRACT

We investigated the extent to which fibroblasts isolated from diverse tissues differ in their capacity to modulate inflammation by comparing the global gene expression profiles of cultured human fibroblasts from skin, acute and chronically inflamed synovium, lymph node and tonsil. The responses of these fibroblasts to TNF-alpha, IFN-gamma and IL-4 stimulation were markedly different, as revealed by hierarchical cluster analysis and principal component analysis. In the absence of exogenous cytokine, synovial and skin fibroblasts exhibited similar patterns of gene expression. However their transcriptional profiles diverged upon treatment with TNF-alpha. This proved to be biologically relevant, as TNF-alpha induced the secretion of different patterns and amounts of IL-6, IL-8 and CCL2 (MCP-1) in the two fibroblast types. Co-culture of skin or synovial fibroblasts with synovial fluid-derived mononuclear cells provided further evidence that these transcriptional differences were functionally significant in an ex vivo setting. Interestingly, the transcriptional response of skin fibroblasts to IL-4 converged with that of TNF-alpha-treated synovial fibroblasts, suggesting resident tissue fibroblasts and their blood-borne precursors may be imprinted by inflammatory cytokines that are characteristic of different tissues. Our data supports the concept that fibroblasts are heterogeneous, and that they contribute to the tissue-specificity of inflammatory reactions. Fibroblasts are therefore likely to play an active role in the persistence of chronic inflammatory reactions.


Subject(s)
Chemokines/biosynthesis , Cytokines/biosynthesis , Fibroblasts/metabolism , Gene Expression Profiling , Adjuvants, Immunologic , Arthritis, Rheumatoid/pathology , Cell Communication/immunology , Chemokines/metabolism , Chemokines/pharmacology , Coculture Techniques , Cytokines/metabolism , Cytokines/pharmacology , Fibroblasts/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Leukocytes, Mononuclear/pathology , Lymph Nodes/pathology , Palatine Tonsil/pathology , Skin/pathology , Synovial Fluid/cytology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...