Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Front Immunol ; 15: 1382638, 2024.
Article in English | MEDLINE | ID: mdl-38715601

ABSTRACT

Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.


Subject(s)
CD4-Positive T-Lymphocytes , CD40 Ligand , Lung , Memory B Cells , Streptococcus pneumoniae , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/metabolism , CD40 Ligand/immunology , Chemokine CXCL13/metabolism , Disease Models, Animal , Immunologic Memory , Lung/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Signal Transduction , Streptococcus pneumoniae/immunology
2.
bioRxiv ; 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36711912

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune disease characterized by progressive multiorgan fibrosis. While the cause of SSc remains unknown, a perturbed vasculature is considered a critical early step in the pathogenesis. Using fibrinogen as a marker of vascular leakage, we found extensive extravascular fibrinogen deposition in the dermis of both limited and diffuse systemic sclerosis disease, and it was present in both early and late-stage patients. Based on a timed series of excision wounds, retention on the fibrin deposit of the splice variant domain, fibrinogen αEC, indicated a recent event, while fibrin networks lacking the αEC domain were older. Application of this timing tool to SSc revealed considerable heterogeneity in αEC domain distribution providing unique insight into disease activity. Intriguingly, the fibrinogen-αEC domain also accumulated in macrophages. These observations indicate that systemic sclerosis is characterized by ongoing vascular leakage resulting in extensive interstitial fibrin deposition that is either continually replenished and/or there is impaired fibrin clearance. Unresolved fibrin deposition might then incite chronic tissue remodeling.

4.
J Clin Invest ; 131(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34060477

ABSTRACT

Lung-resident memory B cells (BRM cells) are elicited after influenza infections of mice, but connections to other pathogens and hosts - as well as their functional significance - have yet to be determined. We postulate that BRM cells are core components of lung immunity. To test this, we examined whether lung BRM cells are elicited by the respiratory pathogen pneumococcus, are present in humans, and are important in pneumonia defense. Lungs of mice that had recovered from pneumococcal infections did not contain organized tertiary lymphoid organs, but did have plasma cells and noncirculating memory B cells. The latter expressed distinctive surface markers (including CD69, PD-L2, CD80, and CD73) and were poised to secrete antibodies upon stimulation. Human lungs also contained B cells with a resident memory phenotype. In mice recovered from pneumococcal pneumonia, depletion of PD-L2+ B cells, including lung BRM cells, diminished bacterial clearance and the level of pneumococcus-reactive antibodies in the lung. These data define lung BRM cells as a common feature of pathogen-experienced lungs and provide direct evidence of a role for these cells in pulmonary antibacterial immunity.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Lung/immunology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/prevention & control , Streptococcus pneumoniae/immunology , Animals , Antigens, Differentiation/immunology , B-Lymphocytes/pathology , Humans , Lung/microbiology , Lung/pathology , Mice , Mice, Transgenic , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology
5.
Ann Rheum Dis ; 79(3): 379-386, 2020 03.
Article in English | MEDLINE | ID: mdl-31767698

ABSTRACT

OBJECTIVES: Determine global skin transcriptome patterns of early diffuse systemic sclerosis (SSc) and how they differ from later disease. METHODS: Skin biopsy RNA from 48 patients in the Prospective Registry for Early Systemic Sclerosis (PRESS) cohort (mean disease duration 1.3 years) and 33 matched healthy controls was examined by next-generation RNA sequencing. Data were analysed for cell type-specific signatures and compared with similarly obtained data from 55 previously biopsied patients in Genetics versus Environment in Scleroderma Outcomes Study cohort with longer disease duration (mean 7.4 years) and their matched controls. Correlations with histological features and clinical course were also evaluated. RESULTS: SSc patients in PRESS had a high prevalence of M2 (96%) and M1 (94%) macrophage and CD8 T cell (65%), CD4 T cell (60%) and B cell (69%) signatures. Immunohistochemical staining of immune cell markers correlated with the gene expression-based immune cell signatures. The prevalence of immune cell signatures in early diffuse SSc patients was higher than in patients with longer disease duration. In the multivariable model, adaptive immune cell signatures were significantly associated with shorter disease duration, while fibroblast and macrophage cell type signatures were associated with higher modified Rodnan Skin Score (mRSS). Immune cell signatures also correlated with skin thickness progression rate prior to biopsy, but did not predict subsequent mRSS progression. CONCLUSIONS: Skin in early diffuse SSc has prominent innate and adaptive immune cell signatures. As a prominently affected end organ, these signatures reflect the preceding rate of disease progression. These findings could have implications in understanding SSc pathogenesis and clinical trial design.


Subject(s)
Adaptive Immunity/genetics , Immunity, Innate/genetics , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/immunology , Adult , Biomarkers/analysis , Biopsy , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Multivariate Analysis , Prospective Studies , Registries , Regression Analysis , Scleroderma, Diffuse/pathology , Sequence Analysis, RNA , Severity of Illness Index , Skin/immunology , Skin/pathology , Transcriptome
6.
J Immunol ; 202(1): 56-68, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30510068

ABSTRACT

Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.


Subject(s)
Dermatitis/immunology , Fibroblasts/physiology , Inflammation/immunology , Lupus Erythematosus, Discoid/immunology , Scleroderma, Systemic/immunology , Skin/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Cells, Cultured , Female , Humans , Male , Middle Aged , Thy-1 Antigens/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Young Adult
7.
Am J Dermatopathol ; 41(1): 16-28, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30320623

ABSTRACT

BACKGROUND: CD90 fibroblasts have been described arising from and replacing the homeostatic CD34 network in scleroderma, but have not been specifically examined in other forms of cutaneous fibrosis. OBJECTIVES: To address expression, timelines, and spatial relationships of CD90, CD34, and smooth muscle actin (SMA) expressing fibroblasts in scars and to examine for the presence of a CD34-to-CD90 transition. METHODS: One hundred and seventeen scars (reparative/hypertrophic/keloidal) were evaluated for CD90, CD34, and SMA expression. Double-staining immunohistochemistry for CD90/CD34 was performed to identify CD90/CD34 transitioning cells, confirmed by double-color immunofluorescence. In addition, some scars were double-stained with CD90/SMA, CD90/procollagen-1, or SMA/procollagen-1 to evaluate spatial relationships and active collagen synthesis. Expression was graded as diffuse, minority, and negative. RESULTS: Most scars demonstrate a CD90/CD34 pattern, and dual CD90/CD34 fibroblasts were observed in 91% of scars. In reparative scars, CD90 expression reverses to a CD34/CD90 state with maturation. Pathologic scars exhibit prolonged CD90 expression. Both CD90 and SMA fibroblasts collagenize scars, although CD90 fibroblasts are more prevalent. CONCLUSIONS: CD90 fibroblasts likely arise from the resting CD34 fibroblastic network. Actively collagenizing scar fibroblasts exhibit a CD90/CD34 phenotype, which is prolonged in pathologic scars. CD90 fibroblasts are likely important players in cutaneous scarring.


Subject(s)
Antigens, CD34/metabolism , Cicatrix, Hypertrophic/metabolism , Fibroblasts/metabolism , Keloid/metabolism , Regeneration , Skin/metabolism , Thy-1 Antigens/metabolism , Actins/metabolism , Biomarkers/metabolism , Cicatrix, Hypertrophic/pathology , Collagen Type I/metabolism , Fibroblasts/pathology , Fibrosis , Fluorescent Antibody Technique , Humans , Keloid/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Procollagen/metabolism , Skin/pathology , Time Factors
8.
Arthritis Rheumatol ; 70(9): 1470-1480, 2018 09.
Article in English | MEDLINE | ID: mdl-29604186

ABSTRACT

OBJECTIVE: To evaluate the clinical efficacy and safety of baminercept, a lymphotoxin ß receptor IgG fusion protein (LTßR-Ig), for the treatment of primary Sjögren's syndrome (SS), and to explore the possible mechanisms of action of this treatment. METHODS: In this multicenter trial, 52 patients with primary SS were randomized in a 2:1 ratio to receive subcutaneous injections of 100 mg of baminercept every week for 24 weeks or matching placebo. The primary end point was the change between screening and week 24 in the stimulated whole salivary flow (SWSF) rate. Secondary end points included the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI), as well as measurements of select chemokines and cytokines and enumeration of peripheral blood B and T cell subsets. RESULTS: The change from baseline to week 24 in the SWSF rate was not significantly different between the baminercept and placebo treatment groups (baseline-adjusted mean change -0.01 versus 0.07 ml/minute; P = 0.332). The change in the ESSDAI during treatment was also not significantly different between the treatment groups (baseline-adjusted mean change -1.23 versus -0.15; P = 0.104). Although the incidence of adverse events was similar between the treatment groups, baminercept therapy was associated with a higher incidence of liver toxicity, including 2 serious adverse events. Baminercept also produced a significant decrease in plasma levels of CXCL13 and significant changes in the number of circulating B and T cells, consistent with its known inhibitory effects on LTßR signaling. CONCLUSION: In this trial, treatment with baminercept failed to significantly improve glandular and extraglandular disease in patients with primary SS, despite evidence from mechanistic studies showing that it blocks LTßR signaling.


Subject(s)
Recombinant Fusion Proteins/therapeutic use , Sjogren's Syndrome/drug therapy , Adult , Aged , B-Lymphocytes/drug effects , Chemokine CXCL13/blood , Double-Blind Method , Female , Humans , Lymphotoxin beta Receptor/immunology , Male , Middle Aged , Recombinant Fusion Proteins/immunology , Sjogren's Syndrome/blood , Sjogren's Syndrome/immunology , T-Lymphocytes/drug effects , Treatment Outcome
9.
Sci Rep ; 7: 41605, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28150703

ABSTRACT

Pulmonary arterial hypertension (PAH) is a fatal condition for which there is no cure. Dimethyl Fumarate (DMF) is an FDA approved anti-oxidative and anti-inflammatory agent with a favorable safety record. The goal of this study was to assess the effectiveness of DMF as a therapy for PAH using patient-derived cells and murine models. We show that DMF treatment is effective in reversing hemodynamic changes, reducing inflammation, oxidative damage, and fibrosis in the experimental models of PAH and lung fibrosis. Our findings indicate that effects of DMF are facilitated by inhibiting pro-inflammatory NFκB, STAT3 and cJUN signaling, as well as ßTRCP-dependent degradation of the pro-fibrogenic mediators Sp1, TAZ and ß-catenin. These results provide a novel insight into the mechanism of its action. Collectively, preclinical results demonstrate beneficial effects of DMF on key molecular pathways contributing to PAH, and support its testing in PAH treatment in patients.


Subject(s)
Dimethyl Fumarate/pharmacology , Hypertension, Pulmonary/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction/drug effects , Animals , Biomarkers , Bleomycin/adverse effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/metabolism , Inflammation/complications , Inflammation/metabolism , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-jun/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , STAT3 Transcription Factor/metabolism
10.
Gut ; 66(8): 1369-1381, 2017 08.
Article in English | MEDLINE | ID: mdl-27196595

ABSTRACT

OBJECTIVE: Lymphotoxin ß receptor (LTßR) signalling has been implicated in inflammation-associated tumour development in different tissues. We have analysed the role of LTßR and alternative NF-κB signalling in Helicobacter pylori-mediated gastric inflammation and pathology. DESIGN: We analysed several ligands and receptors of the alternative NF-κB pathway, RelB, p52 nuclear translocation and target genes in tissue samples of H. pylori-infected patients with different degrees of gastritis or early gastric tumours by in situ hybridisation, immunohistochemistry, Western blot and real-time PCR analyses. Molecular mechanisms involved in LTßR activation by H. pylori were assessed in vitro using human gastric cancer cell lines and distinct H. pylori isolates. The effects of blocking or agonistically activating LTßR on gastric pathology during challenge with a human pathogenic H. pylori strain were studied in a mouse model. RESULTS: Among the tested candidates, LT was significantly increased and activated alternative NF-κB signalling was observed in the gastric mucosa of H. pylori-infected patients. H. pyloriinduced LTßR-ligand expression in a type IV secretion system-dependent but CagA-independent manner, resulting in activation of the alternative NF-κB pathway, which was further enhanced by blocking canonical NF-κB during infection. Blocking LTßR signalling in vivo suppressed H. pylori-driven gastritis, whereas LTßR activation in gastric epithelial cells of infected mice induced a broadened pro-inflammatory chemokine milieu, resulting in exacerbated pathology. CONCLUSIONS: LTßR-triggered activation of alternative NF-κB signalling in gastric epithelial cells executes H. pylori-induced chronic gastritis, representing a novel target to restrict gastric inflammation and pathology elicited by H. pylori, while exclusively targeting canonical NF-κB may aggravate pathology by enhancing the alternative pathway.


Subject(s)
Chemokines/metabolism , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Type IV Secretion Systems/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line, Tumor , Chemokine CCL2/metabolism , Chemokine CCL20/metabolism , Chemokine CXCL10/metabolism , Epithelial Cells/metabolism , Female , Gastric Mucosa/metabolism , Gastritis/microbiology , Helicobacter Infections/complications , Humans , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger , Signal Transduction , Transcription Factor RelB/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
J Clin Invest ; 126(11): 4331-4345, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27721238

ABSTRACT

Scleroderma is a group of skin-fibrosing diseases for which there are no effective treatments. A feature of the skin fibrosis typical of scleroderma is atrophy of the dermal white adipose tissue (DWAT). Adipose tissue contains adipose-derived mesenchymal stromal cells (ADSCs) that have regenerative and reparative functions; however, whether DWAT atrophy in fibrosis is accompanied by ADSC loss is poorly understood, as are the mechanisms that might maintain ADSC survival in fibrotic skin. Here, we have shown that DWAT ADSC numbers were reduced, likely because of cell death, in 2 murine models of scleroderma skin fibrosis. The remaining ADSCs showed a partial dependence on dendritic cells (DCs) for survival. Lymphotoxin ß (LTß) expression in DCs maintained ADSC survival in fibrotic skin by activating an LTß receptor/ß1 integrin (LTßR/ß1 integrin) pathway on ADSCs. Stimulation of LTßR augmented the engraftment of therapeutically injected ADSCs, which was associated with reductions in skin fibrosis and improved skin function. These findings provide insight into the effects of skin fibrosis on DWAT ADSCs, identify a DC-ADSC survival axis in fibrotic skin, and suggest an approach for improving mesenchymal stromal cell therapy in scleroderma and other diseases.


Subject(s)
Dendritic Cells/metabolism , Dermis/metabolism , Scleroderma, Diffuse/metabolism , Subcutaneous Fat/metabolism , Animals , Cell Survival/genetics , Dendritic Cells/pathology , Dermis/pathology , Disease Models, Animal , Female , Fibrosis , Integrin beta1/genetics , Integrin beta1/metabolism , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Mice , Mice, Knockout , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Subcutaneous Fat/pathology
12.
PLoS One ; 11(10): e0164423, 2016.
Article in English | MEDLINE | ID: mdl-27760209

ABSTRACT

Mouse models lupus nephritis (LN) have provided important insights into disease pathogenesis, although none have been able to recapitulate all features of the human disease. Using comprehensive longitudinal analyses, we characterized a novel accelerated mouse model of lupus using pristane treatment in SNF1 (SWR X NZB F1) lupus prone mice (pristane-SNF1 mice). Pristane treatment in SNF1 mice accelerated the onset and progression of proteinuria, autoantibody production, immune complex deposition and development of renal lesions. At week 14, the pristane-SNF1 model recapitulated kidney disease parameters and molecular signatures seen in spontaneous disease in 36 week-old SNF1 mice and in a traditional IFNα-accelerated NZB X NZW F1 (BWF1) model. Blood transcriptome analysis revealed interferon, plasma cell, neutrophil, T-cell and protein synthesis signatures in the pristane-SNF1 model, all known to be present in the human disease. The pristane-SNF1 model appears to be particularly useful for preclinical research, robustly exhibiting many characteristics reminiscent of human disease. These include i) a stronger upregulation of the cytosolic nucleic acid sensing pathway, which is thought to be key component of the pathogenesis of the human disease, and ii) more prominent kidney interstitial inflammation and fibrosis, which have been both associated with poor prognosis in human LN. To our knowledge, this is the only accelerated model of LN that exhibits a robust tubulointerstitial inflammatory and fibrosis response. Taken together our data show that the pristane-SNF1 model is a novel accelerated model of LN with key features similar to human disease.


Subject(s)
Kidney Tubules/drug effects , Kidney Tubules/pathology , Lupus Nephritis/pathology , Terpenes/pharmacology , Animals , Autoantibodies/biosynthesis , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Female , Fibrosis , Glomerulonephritis/chemically induced , Glomerulonephritis/complications , Humans , Hypergammaglobulinemia/chemically induced , Hypergammaglobulinemia/complications , Inflammation/chemically induced , Inflammation/complications , Lupus Nephritis/complications , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Membrane Proteins/metabolism , Mice , Transcription, Genetic/drug effects , Up-Regulation/drug effects
13.
Am J Pathol ; 186(10): 2650-64, 2016 10.
Article in English | MEDLINE | ID: mdl-27565038

ABSTRACT

Tissue injury triggers the activation and differentiation of multiple cell types to minimize damage and initiate repair processes. In systemic sclerosis, these repair processes appear to run unchecked, leading to aberrant remodeling and fibrosis of the skin and multiple internal organs, yet the fundamental pathological defect remains unknown. We describe herein a transition wherein the abundant CD34(+) dermal fibroblasts present in healthy human skin disappear in the skin of systemic sclerosis patients, and CD34(-), podoplanin(+), and CD90(+) fibroblasts appear. This transition is limited to the upper dermis in several inflammatory skin diseases, yet in systemic sclerosis, it can occur in all regions of the dermis. In vitro, primary dermal fibroblasts readily express podoplanin in response to the inflammatory stimuli tumor necrosis factor and IL-1ß. Furthermore, we show that on acute skin injury in both human and murine settings, this transition occurs quickly, consistent with a response to inflammatory signaling. Transitioned fibroblasts partially resemble the cells that form the reticular networks in organized lymphoid tissues, potentially linking two areas of fibroblast research. These results allow for the visualization and quantification of a basic stage of fibroblast differentiation in inflammatory and fibrotic diseases in the skin.


Subject(s)
Fibrosis/pathology , Membrane Glycoproteins/metabolism , Scleroderma, Systemic/pathology , Thy-1 Antigens/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation , Dermis/immunology , Dermis/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Fibrosis/immunology , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Male , Mice , Middle Aged , Scleroderma, Systemic/immunology , Skin/immunology , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism
14.
Kidney Int ; 89(1): 113-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26398497

ABSTRACT

Accumulation of inflammatory cells in different renal compartments is a hallmark of progressive kidney diseases including glomerulonephritis (GN). Lymphotoxin ß receptor (LTßR) signaling is crucial for the formation of lymphoid tissue, and inhibition of LTßR signaling has ameliorated several non-renal inflammatory models. Therefore, we tested whether LTßR signaling could also have a role in renal injury. Renal biopsies from patients with GN were found to express both LTα and LTß ligands, as well as LTßR. The LTßR protein and mRNA were localized to tubular epithelial cells, parietal epithelial cells, crescents, and cells of the glomerular tuft, whereas LTß was found on lymphocytes and tubular epithelial cells. Human tubular epithelial cells, mesangial cells, and mouse parietal epithelial cells expressed both LTα and LTß mRNA upon stimulation with TNF in vitro. Several chemokine mRNAs and proteins were expressed in response to LTßR signaling. Importantly, in a murine lupus model, LTßR blockade improved renal function without the reduction of serum autoantibody titers or glomerular immune complex deposition. Thus, a preclinical mouse model and human studies strongly suggest that LTßR signaling is involved in renal injury and may be a suitable therapeutic target in renal diseases.


Subject(s)
Glomerulonephritis, IGA/metabolism , Lupus Nephritis/metabolism , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/metabolism , RNA, Messenger/analysis , Signal Transduction , Adult , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Female , Glomerulonephritis, IGA/genetics , Humans , Immunoglobulins/pharmacology , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Kidney Tubules/chemistry , Kidney Tubules/metabolism , Kidney Tubules/pathology , Ligands , Lupus Nephritis/genetics , Lymphocytes/chemistry , Lymphotoxin beta Receptor/analysis , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/analysis , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/analysis , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Male , Mesangial Cells/metabolism , Mice , Middle Aged , Signal Transduction/drug effects , Transcriptome
15.
Nat Immunol ; 16(12): 1235-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502405

ABSTRACT

Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphoid Tissue/immunology , Neoplastic Stem Cells/immunology , Stem Cell Niche/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Comparative Genomic Hybridization , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunoblotting , In Situ Hybridization , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Niche/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
16.
Arthritis Rheumatol ; 67(11): 3004-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26240058

ABSTRACT

OBJECTIVE: To define a pharmacodynamic biomarker based on gene expression in skin that would provide a biologic measure of the extent of disease in patients with diffuse cutaneous systemic sclerosis (dcSSc) and could be used to monitor skin disease longitudinally. METHODS: Skin biopsy specimens obtained from a cohort of patients with dcSSc (including longitudinal specimens) were analyzed by microarray. Expression of genes correlating with the modified Rodnan skin thickness score (MRSS) were examined for change over time using a NanoString platform, and a generalized estimating equation (GEE) was used to define and validate longitudinally measured pharmacodynamic biomarkers composed of multiple genes. RESULTS: Microarray analysis of genes parsed to include only those correlating with the MRSS revealed prominent clusters of profibrotic/transforming growth factor ß-regulated, interferon-regulated/proteasome, macrophage, and vascular marker genes. Using genes changing longitudinally with the MRSS, we defined 2 multigene pharmacodynamic biomarkers. The first was defined mathematically by applying a GEE to longitudinal samples. This modeling method selected cross-sectional THBS1 and longitudinal THBS1 and MS4A4A. The second model was based on a weighted selection of genes, including additional genes that changed statistically significantly over time: CTGF, CD163, CCL2, and WIF1. In an independent validation data set, biomarker levels calculated using both models correlated highly with the MRSS. CONCLUSION: Skin gene expression can be used effectively to monitor changes in SSc skin disease over time. We implemented 2 relatively simple models on a NanoString platform permitting highly reproducible assays that can be applied directly to samples from patients or collected as part of clinical trials.


Subject(s)
Scleroderma, Diffuse/pathology , Skin/pathology , Thrombospondin 1 , Antigens/genetics , Biomarkers , Cartilage Oligomeric Matrix Protein/genetics , Cytoskeletal Proteins/genetics , Gene Expression , Humans , Models, Theoretical , Scleroderma, Diffuse/genetics , Severity of Illness Index , Sialic Acid Binding Ig-like Lectin 1/genetics , Thrombospondin 1/genetics
17.
PLoS One ; 9(11): e112545, 2014.
Article in English | MEDLINE | ID: mdl-25405351

ABSTRACT

A subset of patients with autoimmune diseases including rheumatoid arthritis (RA) and lupus appear to be exposed continually to interferon (IFN) as evidenced by elevated expression of IFN induced genes in blood cells. In lupus, detection of endogenous chromatin complexes by the innate sensing machinery is the suspected driver for the IFN, but the actual mechanisms remain unknown in all of these diseases. We investigated in two randomized clinical trials the effects on RA patients of baminercept, a lymphotoxin-beta receptor-immunoglobulin fusion protein that blocks the lymphotoxin-αß/LIGHT axis. Administration of baminercept led to a reduced RNA IFN signature in the blood of patients with elevated baseline signatures. Both RA and SLE patients with a high IFN signature were lymphopenic and lymphocyte counts increased following baminercept treatment of RA patients. These data demonstrate a coupling between the lymphotoxin-LIGHT system and IFN production in rheumatoid arthritis. IFN induced retention of lymphocytes within lymphoid tissues is a likely component of the lymphopenia observed in many autoimmune diseases. ClinicalTrials.gov NCT00664716.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Interferons/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Recombinant Fusion Proteins/therapeutic use , Antirheumatic Agents/adverse effects , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/metabolism , Humans , Interferons/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/pharmacology
18.
Cytokine Growth Factor Rev ; 25(2): 139-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24698108

ABSTRACT

The Lymphotoxin (LT) pathway is best known for its role in orchestrating the development and homeostasis of lymph nodes and Peyer's patches through the regulation of homeostatic chemokines. More recently an appreciation of the LTßR pathway in the production of Type I interferons (IFN-I) during homeostasis and infection has emerged. LTßR signaling is essential in differentiating stromal cells and macrophages in lymphoid organs to rapidly produce IFN-I in response to virus infections independently of the conventional TLR signaling systems. In addition, LTßR signaling is required to produce homeostatic levels of IFN-I from dendritic cells in order to effectively cross-prime a CD8+ T cell response to protein antigen. Importantly, pharmacological inhibition of LTßR signaling in mice has a profound positive impact on a number of autoimmune disease models, although it remains unclear if this efficacy is linked to IFN-I production during chronic inflammation. In this review, we will provide a brief overview of how the "Lymphotoxin Network" is linked to the IFN-I response and its impact on the immune system.


Subject(s)
Autoimmune Diseases/drug therapy , Interferon Type I/immunology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/immunology , Adaptive Immunity , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inflammation/immunology , Interferon Type I/biosynthesis , Macrophages/immunology , Mice , Signal Transduction/immunology
20.
Science ; 343(6176): 1221-8, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24557838

ABSTRACT

Current antiviral agents can control but not eliminate hepatitis B virus (HBV), because HBV establishes a stable nuclear covalently closed circular DNA (cccDNA). Interferon-α treatment can clear HBV but is limited by systemic side effects. We describe how interferon-α can induce specific degradation of the nuclear viral DNA without hepatotoxicity and propose lymphotoxin-ß receptor activation as a therapeutic alternative. Interferon-α and lymphotoxin-ß receptor activation up-regulated APOBEC3A and APOBEC3B cytidine deaminases, respectively, in HBV-infected cells, primary hepatocytes, and human liver needle biopsies. HBV core protein mediated the interaction with nuclear cccDNA, resulting in cytidine deamination, apurinic/apyrimidinic site formation, and finally cccDNA degradation that prevented HBV reactivation. Genomic DNA was not affected. Thus, inducing nuclear deaminases-for example, by lymphotoxin-ß receptor activation-allows the development of new therapeutics that, in combination with existing antivirals, may cure hepatitis B.


Subject(s)
Antiviral Agents/pharmacology , DNA, Circular/metabolism , DNA, Viral/metabolism , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Hepatocytes/drug effects , Interferon-alpha/pharmacology , Lymphotoxin beta Receptor/agonists , Animals , Antibodies, Monoclonal , Antiviral Agents/therapeutic use , Cell Line , Cell Nucleus/virology , Cytidine/metabolism , Cytidine Deaminase/biosynthesis , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon-alpha/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/virology , Lymphotoxin beta Receptor/antagonists & inhibitors , Mice, SCID , Minor Histocompatibility Antigens , Proteins , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...