Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 150(1): 194-206, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770220

ABSTRACT

The differentiation of follicular dendritic cells (FDC) is essential to the remarkable microanatomic plasticity of lymphoid follicles. Here we show that FDC arise from ubiquitous perivascular precursors (preFDC) expressing platelet-derived growth factor receptor ß (PDGFRß). PDGFRß-Cre-driven reporter gene recombination resulted in FDC labeling, whereas conditional ablation of PDGFRß(+)-derived cells abolished FDC, indicating that FDC originate from PDGFRß(+) cells. Lymphotoxin-α-overexpressing prion protein (PrP)(+) kidneys developed PrP(+) FDC after transplantation into PrP(-) mice, confirming that preFDC exist outside lymphoid organs. Adipose tissue-derived PDGFRß(+) stromal-vascular cells responded to FDC maturation factors and, when transplanted into lymphotoxin ß receptor (LTßR)(-) kidney capsules, differentiated into Mfge8(+)CD21/35(+)FcγRIIß(+)PrP(+) FDC capable of trapping immune complexes and recruiting B cells. Spleens of lymphocyte-deficient mice contained perivascular PDGFRß(+) FDC precursors whose expansion required both lymphoid tissue inducer (LTi) cells and lymphotoxin. The ubiquity of preFDC and their strategic location at blood vessels may explain the de novo generation of organized lymphoid tissue at sites of lymphocytic inflammation.


Subject(s)
Blood Vessels/cytology , Dendritic Cells, Follicular/cytology , Spleen/cytology , Stem Cells/cytology , Animals , B-Lymphocytes/immunology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Inflammation/pathology , Killer Cells, Natural/immunology , Mice , Receptor, Platelet-Derived Growth Factor beta/metabolism , Specific Pathogen-Free Organisms , Spleen/metabolism
3.
Nat Immunol ; 13(7): 681-90, 2012 May 27.
Article in English | MEDLINE | ID: mdl-22634865

ABSTRACT

Although cognate encounters between antigen-bearing dendritic cells (DCs) that express the chemokine receptor CCR7 and CCR7(+) naive T cells take place in the T cell zone of lymph nodes, it is unknown whether the colocalization of DCs and T cells in the T cell area is required for the generation of effector cells. Here we found that after infection with an intestinal nematode, antigen-bearing DCs and CD4(+) T cells upregulated the chemokine receptor CXCR5 and localized together outside the T cell zone by a mechanism dependent on the chemokine CXCL13, B cells and lymphotoxin. Notably, lymphotoxin-expressing B cells, CXCR5-expressing DCs and T cells, and CXCL13 were also necessary for development of interleukin 4 (IL-4)-producing type 2 helper T cells (T(H)2 cells), which suggests that T(H)2 differentiation can initiate outside the T cell zone.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphotoxin-alpha/immunology , Receptors, CXCR5/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Chemokine CXCL13/immunology , Interleukin-4/immunology , Lymphocyte Activation/immunology , Lymphotoxin-alpha/biosynthesis , Lymphotoxin-alpha/genetics , Mice , Mice, Inbred C57BL , Nematospiroides dubius/immunology
4.
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
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.
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.
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
9.
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
10.
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
11.
Nat Rev Immunol ; 3(8): 642-55, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12974479

ABSTRACT

Much of the efficiency of the immune system is attributed to the high degree of spatial and temporal organization in the secondary lymphoid organs. Signalling through the lymphotoxin (LT) pathway is a crucial element in the maintenance of this organized microenvironment. The effect of altering lymphoid microenvironments on immune responses remains relatively unexplored. Inhibitors of the LT and LIGHT pathways have been shown to reduce disease in a wide range of autoimmune models. This approach has provided a tool to probe the effect of manipulation of the microenvironment on both normal and pathological immune responses.


Subject(s)
Autoimmune Diseases/immunology , Lymphoid Tissue/immunology , Lymphotoxin-alpha/immunology , Animals , Dendritic Cells, Follicular/immunology , Humans , Lymphotoxin-alpha/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology
12.
Nat Rev Immunol ; 2(7): 465-75, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12094221

ABSTRACT

B-cell-activating factor of the tumour-necrosis-factor family (BAFF) enhances B-cell survival--a function that is indispensable for B-cell maturation--and has a role in enhancing immune responses. Moreover, the overexpression of BAFF results in severe autoimmune disorders in mice, and elevated serum levels of BAFF occur in some patients who have autoimmune diseases. The elucidation of the role of BAFF has set the stage for a new approach to the treatment of autoimmune disease.


Subject(s)
B-Lymphocytes/physiology , Membrane Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Autoimmunity/physiology , B-Cell Activating Factor , B-Cell Activation Factor Receptor , Humans , Models, Molecular , Receptors, Tumor Necrosis Factor/physiology
13.
Blood ; 119(6): 1570-80, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22072556

ABSTRACT

Chronic GVHD (cGVHD) poses a significant risk for HSCT patients. Preclinical development of new therapeutic modalities has been hindered by models with pathologic findings that may not simulate the development of human cGVHD. Previously, we have demonstrated that cGVHD induced by allogeneic HSCT after a conditioning regimen of cyclophosphamide and total-body radiation results in pulmonary dysfunction and airway obliteration, which leads to bronchiolitis obliterans (BO), which is pathognomonic for cGVHD of the lung. We now report cGVHD manifestations in a wide spectrum of target organs, including those with mucosal surfaces. Fibrosis was demonstrated in the lung and liver and was associated with CD4(+) T cells and B220(+) B-cell infiltration and alloantibody deposition. Donor bone marrow obtained from mice incapable of secreting IgG alloantibody resulted in less BO and cGVHD. Robust germinal center reactions were present at the time of cGVHD disease initiation. Blockade of germinal center formation with a lymphotoxin-receptor-immunoglobulin fusion protein suppressed cGVHD and BO. We conclude that cGVHD is caused in part by alloantibody secretion, which is associated with fibrosis and cGVHD manifestations including BO, and that treatment with a lymphotoxin-ß receptor-immunoglobulin fusion protein could be beneficial for cGVHD prevention and therapy.


Subject(s)
B-Lymphocytes/immunology , Bronchiolitis Obliterans/immunology , Germinal Center/immunology , Graft vs Host Disease/immunology , Isoantibodies/immunology , Animals , B-Lymphocytes/metabolism , Blood Donors , Bronchiolitis Obliterans/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chronic Disease , Female , Fluorescent Antibody Technique , Germinal Center/drug effects , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology
14.
Arthritis Rheumatol ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965683

ABSTRACT

OBJECTIVE: Rarefaction of blood and lymphatic vessels in the skin has been reported in SSc (systemic sclerosis, scleroderma). ERG and FLI1 are important regulators of angiogenesis, but their role in lymphatic vasculature is less known. The goal of this study was to determine the role of ERG and FLI1 in postnatal lymphangiogenesis and SSc lymphatic system defects. METHODS: Immunofluorescence was used to detect ERG and FLI1 in SSc and healthy control (HC) skin biopsies. Transcriptional analysis of ERG or FLI1 silenced human dermal lymphatic endothelial cells (LECs) was performed using microarrays. Effects of ERG/FLI1 deficiency on in vitro tubulogenesis in human dermal LECs was examined using a Matrigel assay. Erg and Fli1 endothelial specific knockouts and Erg lymphatic specific knockouts were generated to examine vessel regeneration in mice. RESULTS: ERG and FLI1 protein levels were reduced in the blood and lymphatic vasculature in SSc skin biopsies. ERG was shown to regulate genes involved in lymphatic vessel specification, including VEGFR3/FLT4, LYVE-1, SOX18, and PROX1, while FLI1 enhanced the function of ERG. ERG/FLT4 pathway regulated in vitro tubulogenesis in human LECs. Deficiency of Erg or Fli1 similarly impaired the function of blood vessels in mice. However, only Erg deficiency affected the regeneration of lymphatic vessels during wound healing. CONCLUSION: ERG and FLI1 are essential regulators of blood and lymphatic vessel regeneration. Deficiency of ERG and FLI1 in SSc endothelial cells, may contribute to impairment of blood and lymphatic vasculature in SSc patients.

15.
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
16.
Gastroenterology ; 143(5): 1361-1374, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22863765

ABSTRACT

BACKGROUND & AIMS: Little is known about the pathogenic mechanisms of autoimmune pancreatitis (AIP), an increasingly recognized, immune-mediated form of chronic pancreatitis. Current treatment options are limited and disease relapse is frequent. We investigated factors that contribute to the development of AIP and new therapeutic strategies. METHODS: We used quantitative polymerase chain reaction, immunohistochemical, and enzyme-linked immunosorbent analyses to measure the expression of cytokines and chemokines in tissue and serum samples from patients with and without AIP. We created a mouse model of human AIP by overexpressing lymphotoxin (LT)α and ß specifically in acinar cells (Ela1-LTab mice). RESULTS: Messenger RNA levels of LTα and ß were increased in pancreatic tissues from patients with AIP, compared with controls, and expression of chemokines (CXCL13, CCL19, CCL21, CCL1, and B-cell-activating factor) was increased in pancreatic and serum samples from patients. Up-regulation of these factors was not affected by corticosteroid treatment. Acinar-specific overexpression of LTαß (Ela1-LTαß) in mice led to an autoimmune disorder with various features of AIP. Chronic inflammation developed only in the pancreas but was sufficient to cause systemic autoimmunity. Acinar-specific overexpression of LTαß did not cause autoimmunity in mice without lymphocytes (Ela1-LTab/Rag1(-/-)); moreover, lack of proinflammatory monocytes (Ela1-LTab/Ccr2(-/-)) failed to prevent AIP but prevented early pancreatic tissue damage. Administration of corticosteroids reduced pancreatitis but did not affect production of autoantibodies, such as antipancreatic secretory trypsin inhibitor in Ela1-LTab mice. In contrast, inhibition of LTßR signaling reduced chemokine expression, renal immune-complex deposition, and features of AIP in Ela1-LTab mice. CONCLUSIONS: Overexpression of LTαß specifically in acinar cells of mice causes features of AIP. Reagents that neutralize LTßR ligands might be used to treat patients with AIP.


Subject(s)
Autoimmune Diseases/metabolism , Lymphotoxin beta Receptor/metabolism , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/metabolism , Signal Transduction , Acinar Cells/metabolism , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use , Analysis of Variance , Animals , Autoantibodies/blood , Autoimmune Diseases/blood , Autoimmune Diseases/drug therapy , Case-Control Studies , Cells, Cultured , Chemokines/drug effects , Chemokines/metabolism , Disease Models, Animal , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Lymphocyte Count , Lymphotoxin beta Receptor/blood , Lymphotoxin-alpha/drug effects , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/drug effects , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , Pancreatitis, Chronic/blood , Pancreatitis, Chronic/drug therapy , Promoter Regions, Genetic , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Statistics, Nonparametric , T-Lymphocyte Subsets , Up-Regulation
17.
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.

18.
Immunol Rev ; 223: 202-20, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18613838

ABSTRACT

SUMMARY: The lymphotoxin (LT) system is part of the tumor necrosis factor family and is required for lymph node development. It has provided a wonderful tool for the dissection of processes critical not only for lymphoid organ development but also the maintenance of the adult immune architecture and the formation of ectopic organized lymphoid tissues in chronically inflamed sites. A soluble lymphotoxin-beta receptor-immunoglobulin (LTbetaR-Ig) fusion protein can block this pathway and is currently being tested in the treatment of autoimmune disease. This review focuses on the immunological consequences of combined LT and LIGHT inhibition with LTbetaR-Ig administration as distinct from the developmental biology.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Lymphotoxin alpha1, beta2 Heterotrimer/immunology , Lymphotoxin beta Receptor/immunology , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Movement/drug effects , Cell Movement/immunology , Chemokines/immunology , Clinical Trials as Topic , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Lymphotoxin alpha1, beta2 Heterotrimer/antagonists & inhibitors , Models, Immunological , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Signal Transduction/immunology , Stromal Cells/drug effects , Stromal Cells/immunology
19.
J Biol Chem ; 285(17): 12971-8, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20185819

ABSTRACT

Components of lymphotoxin beta receptor (LTBR)-associated signaling complexes, including TRAF2, TRAF3, NIK, IKK1, and IKK2 have been shown to participate in the coupling of LTBR to NFkappaB. Here, we report that TRAF3 functions as a negative regulator of LTBR signaling via both canonical and non-canonical NFkappaB pathways by two distinct mechanisms. Analysis of NFkappaB signaling in cell lines with functionally intact NFkappaB pathway but lacking LTBR-mediated induction of NFkappaB target genes revealed an inverse association of cellular TRAF3 levels with LTBR-specific defect in canonical NFkappaB activation. Increased expression of TRAF3 correlated with its increased recruitment to LTBR-induced signaling complexes, decreased recruitment of TRAF2, and attenuated phosphorylation of IkappaB alpha and RelA. In contrast, activation of NFkappaB by TNF did not depend on TRAF3 levels. siRNA-mediated depletion of TRAF3 promoted recruitment of TRAF2 and IKK1 to activated LTBR, enabling LTBR-inducible canonical NFkappaB signaling and NFkappaB target gene expression. TRAF3 knock-down also increased mRNA and protein expression of several non-canonical NFkappaB components, including NFkappaB2/p100, RelB, and NIK, accompanied by processing of NFkappaB2/p100 into p52. These effects of TRAF3 depletion did not require LTBR signaling and were consistent with autonomous activation of the non-canonical NFkappaB pathway. Our data illustrate the function of TRAF3 as a dual-mode repressor of LTBR signaling that controls activation of canonical NFkappaB, and de-repression of the intrinsic activity of non-canonical NFkappaB. Modulation of cellular TRAF3 levels may thus contribute to regulation of NFkappaB-dependent gene expression by LTBR by affecting the balance of LTBR-dependent activation of canonical and non-canonical NFkappaB pathways.


Subject(s)
Gene Expression Regulation/physiology , Lymphotoxin beta Receptor/metabolism , NF-kappa B p52 Subunit/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Lymphotoxin beta Receptor/genetics , NF-kappa B p52 Subunit/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/genetics , Transcription Factor RelA/genetics , Transcription Factor RelB/genetics , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL