Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
J Clin Invest ; 127(4): 1517-1530, 2017 Apr 03.
Article En | MEDLINE | ID: mdl-28240604

Tissue fibrosis is the primary cause of long-term graft failure after organ transplantation. In lung allografts, progressive terminal airway fibrosis leads to an irreversible decline in lung function termed bronchiolitis obliterans syndrome (BOS). Here, we have identified an autocrine pathway linking nuclear factor of activated T cells 2 (NFAT1), autotaxin (ATX), lysophosphatidic acid (LPA), and ß-catenin that contributes to progression of fibrosis in lung allografts. Mesenchymal cells (MCs) derived from fibrotic lung allografts (BOS MCs) demonstrated constitutive nuclear ß-catenin expression that was dependent on autocrine ATX secretion and LPA signaling. We found that NFAT1 upstream of ATX regulated expression of ATX as well as ß-catenin. Silencing NFAT1 in BOS MCs suppressed ATX expression, and sustained overexpression of NFAT1 increased ATX expression and activity in non-fibrotic MCs. LPA signaling induced NFAT1 nuclear translocation, suggesting that autocrine LPA synthesis promotes NFAT1 transcriptional activation and ATX secretion in a positive feedback loop. In an in vivo mouse orthotopic lung transplant model of BOS, antagonism of the LPA receptor (LPA1) or ATX inhibition decreased allograft fibrosis and was associated with lower active ß-catenin and dephosphorylated NFAT1 expression. Lung allografts from ß-catenin reporter mice demonstrated reduced ß-catenin transcriptional activation in the presence of LPA1 antagonist, confirming an in vivo role for LPA signaling in ß-catenin activation.


Autocrine Communication , Lysophospholipids/physiology , Pulmonary Fibrosis/metabolism , beta Catenin/metabolism , Allografts , Animals , Bronchiolitis Obliterans/metabolism , Cells, Cultured , Collagen/biosynthesis , Female , Graft Rejection/metabolism , Humans , Lung/metabolism , Lung/pathology , Lung Transplantation , Male , Mice, Inbred C57BL , Mice, Inbred DBA , NFATC Transcription Factors/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Transcriptional Activation
2.
JCI Insight ; 2(1): e89504, 2017 01 12.
Article En | MEDLINE | ID: mdl-28097233

Mechanical complications of myocardial infarction (MI) are often fatal. Little is known about endogenous factors that predispose to myocardial rupture after MI. Ectonucleoside triphosphate diphosphohydrolase (CD39) could be a critical mediator of propensity to myocardial rupture after MI due to its role in modulating inflammation and thrombosis. Using a model of permanent coronary artery ligation, rupture was virtually abrogated in cd39-/- mice versus cd39+/+ controls, with elevated fibrin and collagen deposition and marked neutrophil and macrophage influx. Macrophages were found to display increased surface expression of CD301 and CD206, marking a reparative phenotype, driven by increased extracellular ATP and IL-4 in the infarcted myocardium of cd39-/- mice. A myeloid-specific CD39-knockout mouse also demonstrated protection from rupture, with an attenuated rupture phenotype, suggesting that complete ablation of CD39 provides the greatest degree of protection in this model. Absence of CD39, either globally or in a myeloid lineage-restricted fashion, skews the phenotype toward alternatively activated (reparative) macrophage infiltration following MI. These studies reveal a previously unrecognized and unexpected role of endogenous CD39 to skew macrophage phenotype and promote a propensity to myocardial rupture after MI.


Adenosine Triphosphatases/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Rupture/etiology , Animals , Antigens, CD/metabolism , Disease Models, Animal , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Phenotype , Thrombosis/metabolism
3.
Stem Cells ; 30(9): 2010-9, 2012 Sep.
Article En | MEDLINE | ID: mdl-22782863

Mesenchymal stem cells (MSCs) have been demonstrated to reside in human adult organs. However, mechanisms of migration of these endogenous MSCs within their tissue of origin are not well understood. Here, we investigate migration of human adult lung-resident (LR) mesenchymal progenitor cells. We demonstrate that bioactive lipid lysophosphatidic acid (LPA) plays a principal role in the migration of human LR-MSCs through a signaling pathway involving LPA1-induced ß-catenin activation. LR-MSCs isolated from human lung allografts and lungs of patients with scleroderma demonstrated a robust migratory response to LPA in vitro. Furthermore, LPA levels correlated with LR-MSC numbers in bronchoalveolar lavage (BAL), providing demonstration of the in vivo activity of LPA in human adult lungs. Migration of LR-MSCs was mediated via LPA1 receptor ligation and LPA1 silencing significantly abrogated the migratory response of LR-MSCs to LPA as well as human BAL. LPA treatment of LR-MSCs induced protein kinase C-mediated glycogen synthase kinase-3ß phosphorylation, with resulting cytoplasmic accumulation and nuclear translocation of ß-catenin. TCF/LEF dual luciferase gene reporter assay demonstrated a significant increase in transcriptional activity after LPA treatment. LR-MSC migration and increase in reporter gene activity in the presence of LPA were abolished by transfection with ß-catenin small interfering RNA demonstrating that ß-catenin is critical in mediating LPA-induced LR-MSC migration. These data delineate a novel signaling pathway through which ligation of a G protein-coupled receptor by a biologically relevant lipid mediator induces migration of human tissue-resident mesenchymal progenitors.


Cell Movement/drug effects , Lysophospholipids/pharmacology , Mesenchymal Stem Cells/cytology , beta Catenin/metabolism , Cell Movement/physiology , Cells, Cultured , Humans , Lung/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Transfection , beta Catenin/genetics
4.
Am J Respir Crit Care Med ; 185(1): 77-84, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-21940790

RATIONALE: Donor mesenchymal stromal/stem cell (MSC) expansion and fibrotic differentiation is associated with development of bronchiolitis obliterans syndrome (BOS) in human lung allografts. However, the regulators of fibrotic differentiation of these resident mesenchymal cells are not well understood. OBJECTIVES: This study examines the role of endogenous and exogenous prostaglandin (PG)E2 as a modulator of fibrotic differentiation of human lung allograft-derived MSCs. METHODS: Effect of PGE2 on proliferation, collagen secretion, and α-smooth muscle actin (α-SMA) expression was assessed in lung-resident MSCs (LR-MSCs) derived from patients with and without BOS. The response pathway involved was elucidated by use of specific agonists and antagonists. MEASUREMENT AND MAIN RESULTS: PGE2 treatment of LR-MSCs derived from normal lung allografts significantly inhibited their proliferation, collagen secretion, and α-SMA expression. On the basis of pharmacologic and small-interfering RNA approaches, a PGE2/E prostanoid (EP)2/adenylate cyclase pathway was implicated in these suppressive effects. Stimulation of endogenous PGE2 secretion by IL-1ß was associated with amelioration of their myofibroblast differentiation in vitro, whereas its inhibition by indomethacin augmented α-SMA expression. LR-MSCs from patients with BOS secreted significantly less PGE2 than non-BOS LR-MSCs. Furthermore, BOS LR-MSCs were found to be defective in their ability to induce cyclooxygenase-2, and therefore unable to up-regulate PGE2 synthesis in response to IL-1ß. BOS LR-MSCs also demonstrated resistance to the inhibitory actions of PGE2 in association with a reduction in the EP2/EP1 ratio. CONCLUSIONS: These data identify the PGE2 axis as an important autocrine-paracrine brake on fibrotic differentiation of LR-MSCs, a failure of which is associated with BOS.


Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/pathology , Dinoprostone/metabolism , Lung Transplantation , Lung/metabolism , Lung/pathology , Actins/metabolism , Adenylyl Cyclases/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid , Cell Differentiation , Cells, Cultured , Collagen/metabolism , Cyclooxygenase 2/metabolism , Fibrosis , Humans , Interleukin-1beta/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Real-Time Polymerase Chain Reaction
5.
Am J Pathol ; 178(6): 2461-9, 2011 Jun.
Article En | MEDLINE | ID: mdl-21641374

Fibrotic obliteration of the small airways leading to progressive airflow obstruction, termed bronchiolitis obliterans syndrome (BOS), is the major cause of poor outcomes after lung transplantation. We recently demonstrated that a donor-derived population of multipotent mesenchymal stem cells (MSCs) can be isolated from the bronchoalveolar lavage (BAL) fluid of human lung transplant recipients. Herein, we study the organ specificity of these cells and investigate the role of local mesenchymal progenitors in fibrogenesis after lung transplantation. We demonstrate that human lung allograft-derived MSCs uniquely express embryonic lung mesenchyme-associated transcription factors with a 35,000-fold higher expression of forkhead/winged helix transcription factor forkhead box (FOXF1) noted in lung compared with bone marrow MSCs. Fibrotic differentiation of MSCs isolated from normal lung allografts was noted in the presence of profibrotic mediators associated with BOS, including transforming growth factor-ß and IL-13. MSCs isolated from patients with BOS demonstrated increased expression of α-SMA and collagen I when compared with non-BOS controls, consistent with a stable in vivo fibrotic phenotype. FOXF1 mRNA expression in the BAL cell pellet correlated with the number of MSCs in the BAL fluid, and myofibroblasts present in the fibrotic lesions expressed FOXF1 by in situ hybridization. These data suggest a key role for local tissue-specific, organ-resident, mesenchymal precursors in the fibrogenic processes in human adult lungs.


Lung Transplantation , Lung/pathology , Mesenchymal Stem Cells/pathology , Actins/metabolism , Biomarkers/metabolism , Biopsy , Bone Marrow Cells/pathology , Bronchiolitis Obliterans/pathology , Bronchoalveolar Lavage Fluid , Cell Count , Cell Differentiation , Cell Separation , Collagen/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Lung/embryology , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Myofibroblasts/pathology , Organ Specificity , Phenotype , Receptors, Interleukin-13/metabolism , Transplantation, Homologous
6.
Am J Respir Cell Mol Biol ; 45(4): 809-16, 2011 Oct.
Article En | MEDLINE | ID: mdl-21378261

Multipotent mesenchymal progenitor cells, termed "mesenchymal stem cells" (MSCs), have been demonstrated to reside in human adult lungs. However, there is little information regarding the associations of these local mesenchymal progenitors with other resident somatic cells and their potential for therapeutic use. Here we provide in vivo and in vitro evidence for the ability of human adult lung-resident MSCs (LR-MSCs) to interact with the local epithelial cells. The in vivo retention and localization of human LR-MSCs in an alveolar microenvironment was investigated by placing PKH-26 or DsRed lentivirus-labeled human LR-MSCs in the lungs of immunodeficient (SCID) mice. At 3 weeks after intratracheal administration, 19.3 ± 3.21% of LR-MSCs were recovered, compared with 3.47 ± 0.51% of control fibroblasts, as determined by flow cytometry. LR-MSCs were found to persist in murine lungs for up to 6 months and demonstrated preferential localization to the corners of the alveoli in close proximity to type II alveolar epithelial cells, the progenitor cells of the alveolar epithelium. In vitro, LR-MSCs established gap junction communications with lung alveolar and bronchial epithelial cells and demonstrated an ability to secrete keratinocyte growth factor, an important modulator of epithelial cell proliferation and differentiation. Gap junction communications were also demonstrable between LR-MSCs and resident murine cells in vivo. This study demonstrates, for the first time, an ability of tissue-specific MSCs to engraft in their organ of origin and establishes a pathway of bidirectional interaction between these mesenchymal progenitors and adult somatic epithelial cells in the lung.


Alveolar Epithelial Cells/metabolism , Cell Communication , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Pulmonary Alveoli/surgery , Animals , Cell Separation/methods , Cell Tracking , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 7/metabolism , Flow Cytometry , Fluorescent Dyes/metabolism , Gap Junctions/metabolism , Genetic Vectors , Graft Survival , Humans , Lentivirus/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, SCID , Organic Chemicals/metabolism , Pulmonary Alveoli/metabolism , Time Factors , Transfection , Red Fluorescent Protein
7.
Am J Respir Crit Care Med ; 183(8): 1062-70, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21169468

RATIONALE: Bronchoalveolar lavage fluid (BAL) from human lung allografts demonstrates the presence of a multipotent mesenchymal stromal cell population. However, the clinical relevance of this novel cellular component of BAL and its association with bronchiolitis obliterans syndrome (BOS), a disease marked by progressive airflow limitation secondary to fibrotic obliteration of the small airways, remains to be determined. OBJECTIVES: In this study we investigate the association of number of mesenchymal stromal cells in BAL with development of BOS in human lung transplant recipients. METHODS: Mesenchymal colony-forming units (CFUs) were quantitated in a cohort of 405 BAL samples obtained from 162 lung transplant recipients. Poisson generalized estimating equations were used to determine the predictors of BAL mesenchymal CFU count. MEASUREMENTS AND MAIN RESULTS: Higher CFU counts were noted early post-transplantation; time from transplant to BAL of greater than 3 months predicted 0.4-fold lower CFU counts (P = 0.0001). BOS diagnosis less than or equal to 365 days before BAL was associated with a 2.11-fold higher CFU count (P = 0.02). There were 2.62- and 2.70-fold higher CFU counts noted in the presence of histologic diagnosis of bronchiolitis obliterans (P = 0.05) and organizing pneumonia (0.0003), respectively. In BAL samples obtained from BOS-free patients greater than 6 months post-transplantation (n = 173), higher mesenchymal CFU counts (≥10) significantly predicted BOS onset in both univariate (hazard ratio, 5.61; 95% CI, 3.03-10.38; P < 0.0001) and multivariate (hazard ratio, 5.02; 95% CI, 2.40-10.51; P < 0.0001) Cox regression analysis. CONCLUSIONS: Measurement of mesenchymal CFUs in the BAL provides predictive information regarding future BOS onset.


Bronchiolitis Obliterans/etiology , Bronchoalveolar Lavage Fluid/cytology , Lung Transplantation/adverse effects , Mesenchymal Stem Cells/physiology , Adult , Aged , Biomarkers , Female , Flow Cytometry , Humans , Male , Middle Aged , Poisson Distribution , Predictive Value of Tests , Proportional Hazards Models , Statistics, Nonparametric , Stem Cells/cytology , Young Adult
8.
J Immunol ; 181(6): 4389-96, 2008 Sep 15.
Article En | MEDLINE | ID: mdl-18768898

Development of allograft rejection continues to be the major determinant of morbidity and mortality postlung transplantation. We have recently demonstrated that a population of donor-derived mesenchymal stem cells is present in human lung allografts and can be isolated and expanded ex vivo. In this study, we investigated the impact of lung resident mesenchymal stem cells (LR-MSCs), derived from allografts of human lung transplant recipients, on T cell activation in vitro. Similar to bone marrow-derived MSCs, LR-MSCs did not express MHC II or the costimulatory molecules CD80 or CD86. In vitro, LR-MSCs profoundly suppressed the proliferative capacity of T cells in response to a mitogenic or an allogeneic stimulus. The immunosuppressive function of LR-MSCs was also noted in the absence of direct cell contact, indicating that LR-MSCs mediated their effect predominantly via a soluble mediator. LR-MSCs isolated from lung transplant recipients demonstrated PGE(2) secretion at baseline (385 +/- 375 pg/ml), which increased in response to IL-1beta (1149 +/- 1081 pg/ml). The addition of PG synthesis inhibitors (indomethacin and NS-398) substantially abrogated LR-MSC-mediated immunosuppression, indicating that PGE(2) may be one of the major soluble mediators impacting T cell activity. This is the first report to demonstrate that human tissue-derived MSCs isolated from an allogeneic environment have the potential to mediate immunological responses in vitro.


Cell Proliferation , Dinoprostone/physiology , Growth Inhibitors/physiology , Immune Tolerance , Lung Transplantation/immunology , Lung/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocyte Subsets/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Dinoprostone/biosynthesis , Growth Inhibitors/biosynthesis , Humans , Lung/cytology , Lung/metabolism , Mesenchymal Stem Cells/metabolism , T-Lymphocyte Subsets/immunology , Transplantation, Homologous
9.
J Clin Invest ; 117(4): 989-96, 2007 Apr.
Article En | MEDLINE | ID: mdl-17347686

The origin and turnover of connective tissue cells in adult human organs, including the lung, are not well understood. Here, studies of cells derived from human lung allografts demonstrate the presence of a multipotent mesenchymal cell population, which is locally resident in the human adult lung and has extended life span in vivo. Examination of plastic-adherent cell populations in bronchoalveolar lavage samples obtained from 76 human lung transplant recipients revealed clonal proliferation of fibroblast-like cells in 62% (106 of 172) of samples. Immunophenotyping of these isolated cells demonstrated expression of vimentin and prolyl-4-hydroxylase, indicating a mesenchymal phenotype. Multiparametric flow cytometric analyses revealed expression of cell-surface proteins, CD73, CD90, and CD105, commonly found on mesenchymal stem cells (MSCs). Hematopoietic lineage markers CD14, CD34, and CD45 were absent. Multipotency of these cells was demonstrated by their capacity to differentiate into adipocytes, chondrocytes, and osteocytes. Cytogenetic analysis of cells from 7 sex-mismatched lung transplant recipients harvested up to 11 years after transplant revealed that 97.2% +/- 2.1% expressed the sex genotype of the donor. The presence of MSCs of donor sex identity in lung allografts even years after transplantation provides what we believe to be the first evidence for connective tissue cell progenitors that reside locally within a postnatal, nonhematopoietic organ.


Lung Transplantation/physiology , Mesenchymal Stem Cells/cytology , Adult , Antigens, CD/analysis , Bronchoalveolar Lavage Fluid/cytology , Connective Tissue Cells/cytology , Connective Tissue Cells/physiology , Female , Flow Cytometry , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Lung Diseases/classification , Lung Diseases/surgery , Male , Mesenchymal Stem Cells/physiology , Middle Aged , Transplantation, Homologous
10.
Am J Physiol Lung Cell Mol Physiol ; 293(1): L124-30, 2007 Jul.
Article En | MEDLINE | ID: mdl-17384085

Chronic airway rejection, characterized by lymphocytic bronchitis, epithelial cell damage, and obliterative bronchiolitis (OB), limits long-term survival after lung transplantation. The transcription factor early growth response gene-1 (Egr-1) induces diverse inflammatory mediators, some involved in OB pathogenesis. An orthotopic mouse tracheal transplant model was used to determine whether Egr-1 promotes development of airway allograft rejection. Significantly higher Egr-1 mRNA levels were seen in allografts (3.2-fold increase vs. isografts, P = 0.012). Allografts revealed thickening of epithelial and subepithelial airway layers (51 +/- 4% luminal encroachment for allografts vs. 20 +/- 3% for isografts, P < 0.0001) marked by significant lymphocytic infiltration. Absence of the Egr-1 gene in donor (but not recipient) tissue resulted in significant reduction in luminal narrowing (34 +/- 4%, P = 0.0001) with corresponding diminution of T cell infiltration. Egr-1 null allografts exhibited a striking reduction in inducible nitric oxide synthase (iNOS) expression. Effector cytokines previously implicated in OB pathogenesis with known Egr-1 promoter motifs (IL-1beta and JE/monocyte chemoattractant protein-1) were reduced in Egr-1 null allografts. These data suggest a paradigm wherein local induction of Egr-1 in tracheal allografts drives expression of inflammatory mediators responsible for lymphocyte recruitment and tissue destruction characteristic of airway rejection.


Early Growth Response Protein 1/genetics , Graft Rejection/metabolism , Lung Transplantation , Animals , CD3 Complex/metabolism , Cytokines/blood , Early Growth Response Protein 1/metabolism , Gene Expression Regulation , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transplantation, Homologous
11.
Am J Pathol ; 169(1): 47-60, 2006 Jul.
Article En | MEDLINE | ID: mdl-16816360

The pathogenesis of bronchiolitis obliterans (BO), a common and devastating obliterative disorder of small airways following lung transplantation, remains poorly understood. Lesions are characterized in their early stages by lymphocyte influx that evolves into dense fibrotic infiltrates. Airway specimens taken from patients with histological BO revealed infiltrating myofibroblasts, which strongly expressed the signaling chain of the high affinity interleukin-13 (IL-13) receptor IL-13Ralpha1. Because IL-13 has proinflammatory and profibrotic actions, a contributory role for IL-13 in BO development was examined using murine models of orthotopic and heterotopic tracheal transplantation. Compared with airway isografts, allografts exhibited a significant increase in relative IL-13 mRNA and protein levels. Allogeneic tracheas transplanted into IL-13-deficient mice were protected from BO in both transplant models. Flow cytometric analysis of orthotopic transplant tissue digests revealed markedly fewer infiltrating mononuclear phagocytes and CD3(+) T lymphocytes in IL-13-deficient recipients. Furthermore, protection from luminal obliteration, collagen deposition, and myofibroblast infiltration was observed in heterotopic airways transplanted into the IL-13(-/-) recipients. Transforming growth factor-beta1 expression was significantly decreased in tracheal allografts into IL-13(-/-) recipients, compared to wild-type counterparts. These human and murine data implicate IL-13 as a critical effector cytokine driving cellular recruitment and subsequent fibrosis in clinical and ex-perimental BO.


Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/pathology , Interleukin-13/metabolism , Trachea/pathology , Adult , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Flow Cytometry , Humans , Interleukin-13 Receptor alpha1 Subunit , Male , Mice , Middle Aged , Polymerase Chain Reaction , Proteins/metabolism , RNA, Messenger/analysis , Receptors, Interleukin/metabolism , Receptors, Interleukin-13 , Trachea/immunology , Trachea/transplantation , Transplantation, Homologous , Transplantation, Isogeneic
12.
J Interferon Cytokine Res ; 25(10): 587-94, 2005 Oct.
Article En | MEDLINE | ID: mdl-16241857

Interferon-gamma (IFN-gamma) is a pleiotropic cytokine with potent antitumor effects, both in vitro and in vivo. The antitumor activity of IFN-gamma is mediated in part through IFN regulatory factor-1 (IRF-1) and may be blocked by IRF-2. To test our hypothesis that some tumors escape the antitumor effects of IFN-gamma by cellular changes reflected in IRF-1 and IRF-2 expression, we examined IRF-1 and IRF-2 expression in tissue microarrays (TMA) containing 187 specimens of clinically defined invasive breast carcinoma. TMAs (Cooperative Breast Cancer Tissue Resource [CBCTR], National Cancer Institute [NCI]) were stained and then scored by three evaluators blinded to the patients' clinical status. After final scoring, the CBCTR provided the available clinical data for each patient. Whether sorted by carcinoma type or for all data together, statistical analysis showed a significant positive correlation between IRF-1 and IRF-2 expression (p = 0.01) and a negative correlation between IRF-1 expression and tumor grade (p = 0.005). IRF-1 expression is consistent with its role as a tumor suppressor; high-grade breast carcinomas were less likely to maintain expression of IRF-1, a finding consistent with a role for IRF-1 as a tumor suppressor. Further, tumors maintained expression of IRF-2 if there was coincident expression of IRF-1. These data support a model in which alterations of the expression of intracellular effectors of IFN-gamma signaling may diminish the immune-mediated tumor control mechanisms of IFN-gamma.


Breast Neoplasms/metabolism , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-2/biosynthesis , Tissue Array Analysis , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinoma/immunology , Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-2/immunology , Interferon-gamma/immunology , Signal Transduction/immunology , Tissue Array Analysis/methods , Tumor Escape/immunology
...