Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Arthritis Res Ther ; 23(1): 246, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34560894

ABSTRACT

BACKGROUND: In patients with axial spondyloarthritis (axSpA), monocytes show a pre-activated phenotype. Gut inflammation is a trigger of monocyte activation and may also affect their development in the bone marrow (BM). As gut inflammation is commonly observed in axSpA patients, we performed a detailed analysis of monocyte transcriptomes of axSpA patients in two cohorts and searched for signs of activation and developmental adaptations as putative imprints of gut inflammation. METHODS: Transcriptomes of blood CD14+ monocytes of HLA-B27+ axSpA patients and healthy controls (HC) were generated by microarrays from cohort 1 and by RNA-sequencing from cohort 2. Differentially expressed genes from both analyses were subjected to gene set enrichment analysis (GSEA) and to co-expression analysis in reference transcriptomes from BM cells, blood cells and activated monocytes. As serological markers of translocation, 1,3 beta-glycan, intestinal fatty acid binding protein, and lipopolysaccharide binding protein (LBP) were determined by LAL and ELISA. RESULTS: Transcriptome analysis identified axSpA-specific monocyte signatures showing an imprint of LPS/cytokine-activated monocytes, late granulopoietic BM cells, blood neutrophils, and G-CSF-mobilized blood cells, which suggests LPS/TNF activation and more prominent BM adaptation promoting a neutrophil-like phenotype. GSEA mapped axSpA upregulated genes to inflammatory responses and TNFα signaling and downregulated probe-sets to metabolic pathways. Among translocation markers, LBP levels were significantly increased in axSpA patients vs. HC (p < 0.001). Stratified analysis by disease activity and stage identified an "active disease signature" (BASDAI ≥ 4) with an imprint of LPS/cytokine-activated monocytes and CD16+ monocyte subsets. The "AS signature" (vs. non-radiographic axSpA) showed a reinforced neutrophil-like phenotype due to deprivation of dendritic cell transcripts. CONCLUSIONS: The neutrophil-like phenotype of axSpA monocytes points towards a biased monocytopoiesis from granulocyte-monocyte progenitors. This shift in monocytopoiesis and the LPS/cytokine imprint as well as the elevated LBP levels are indicators of systemic inflammation, which may result from bacterial translocation. The BM adaptation is most prominent in AS patients while disease activity appears to be linked to activation and trafficking of monocytes.


Subject(s)
Monocytes , Spondylarthritis , Cytokines , Gene Expression Profiling , Humans , Spondylarthritis/genetics , Transcriptome
2.
PLoS One ; 16(9): e0254557, 2021.
Article in English | MEDLINE | ID: mdl-34473704

ABSTRACT

The interaction of extracellular matrix (ECM) components with hepatic stellate cells (HSCs) is thought to perpetuate fibrosis by stimulating signaling pathways that drive HSC activation, survival and proliferation. Consequently, disrupting the interaction between ECM and HSCs is considered a therapeutical avenue although respective targets and underlying mechanisms remain to be established. Here we have interrogated the interaction between type VI collagen (CVI) and HSCs based on the observation that CVI is 10-fold upregulated during fibrosis, closely associates with HSCs in vivo and promotes cell proliferation and cell survival in cancer cell lines. We exposed primary rat HSCs and a rat hepatic stellate cell line (CFSC) to soluble CVI and determined the rate of proliferation, apoptosis and fibrogenesis in the absence of any additional growth factors. We find that CVI in nanomolar concentrations prevents serum starvation-induced apoptosis. This potent anti-apoptotic effect is accompanied by induction of proliferation and acquisition of a pronounced pro-fibrogenic phenotype characterized by increased α-smooth muscle actin, TGF-ß, collagen type I and TIMP-1 expression and diminished proteolytic MMP-13 expression. The CVI-HSC interaction can be disrupted with the monomeric α2(VI) and α3(VI) chains and abrogates the activating CVI effects. Further, functional relevant α3(VI)-derived 30 amino acid peptides lead to near-complete inhibition of the CVI effect. In conclusion, CVI serves as a potent mitogen and activating factor for HSCs. The antagonistic effects of the CVI monomeric chains and peptides point to linear peptide sequences that prevent activation of CVI receptors which may allow a targeted antifibrotic therapy.


Subject(s)
Collagen Type VI/metabolism , Fibrosis/drug therapy , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Peptides/pharmacology , Protein Subunits/pharmacology , Transforming Growth Factor beta1/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibrosis/metabolism , Fibrosis/pathology , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Rats , Signal Transduction
3.
Biomed Pharmacother ; 130: 110587, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32763819

ABSTRACT

Doxorubicin (DOX) is commonly used as an anti-cancer agent. However, its severe cardiotoxicity often makes it life threatening even long after DOX therapy during childhood. We recently reported interferon-γ (IFN-γ) necessary for DOX-induced acute cardiotoxicity in a p38 dependent way and, asked here for the potential of IFN-γ blockade to prevent DOX-induced chronic cardiotoxicity during tumor therapy. In our model system, mice without or with growing tumors repeatedly received DOX treatment. Simultaneous injection of anti-IFN-γ antibody R46-A2 with DOX to block IFN-γ signal efficiently protected the cardiac function of DOX treated recipients. Importantly, a single late injection of R46-A2 after DOX exposure also ameliorated DOX induced cardiac dysfunction in tumor-bearing mice. The anti-IFN-γ treatment did not affect the DOX-mediated tumor suppression effect and it left the main cellular immune response intact. Therefore, temporary blockade of IFN-γ may represent a novel strategy to ameliorate established DOX induced cardiotoxicity (DIC) or prevent its development in tumor therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies/therapeutic use , Cardiotoxicity/drug therapy , Doxorubicin/therapeutic use , Interferon-gamma/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/adverse effects , Cell Line, Tumor , Doxorubicin/adverse effects , Female , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Crohns Colitis ; 13(1): 79-91, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30272118

ABSTRACT

BACKGROUND AND AIMS: Creeping fat [CF] is a hyperplasia of adipose tissue adjacent to inflamed intestine in Crohn's disease [CD]. Data from genome-wide association studies [GWAS] distinguished Crohn's colitis from ileal CD and ulcerative colitis [UC]. This study analysed the T-cell compartments of ileal and colonic mesenteric fat and corresponding mucosa to provide cellular proof for the suggested GWAS classification. METHODS: Samples were obtained from 34 CD or UC patients. Cells were analysed by immunohistochemistry and flow cytometry, and tissue cytokine release was assessed by cytometric bead array. RESULTS: Only ileal CF revealed the distinct adipocyte hyperplasia combined with dense T-cell infiltration and fibrosis; colonic fat from CD and UC patients lacked these findings. T-cell subpopulations differed between mesenteric fat in ileal CD, colonic CD and UC: ileal CF had nearly 10 times more T-cells than colonic fat. The proportions of regulatory and central memory T-cells were significantly higher in ileal CF compared with colonic fat in CD and UC. In all groups, the mucosal T-cell compartment was distinct from the mesenteric fat. Remarkably, correlation between disease activity and proportion of pro- and anti-inflammatory T-cell subpopulations was inverse, comparing ileal and colonic fat in CD. CONCLUSIONS: This first in-depth analysis of the T-cell compartment in ileal and colonic mesenteric adipose tissue in CD and UC identifies a unique T-cell niche in the ileal mesenteric fat tissue in CD. From a clinical point of view, our findings underscore the novel concept of colonic and ileal CD as distinct IBD entities.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Intestinal Mucosa/immunology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , T-Lymphocytes , Adult , Aged , CD4-CD8 Ratio , Cadherins/metabolism , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon , Crohn Disease/genetics , Crohn Disease/pathology , Cytokines/metabolism , Female , Fibrosis , Genome-Wide Association Study , Humans , Hyperplasia/pathology , Ileum , Integrins/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Severity of Illness Index , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Young Adult
5.
J Pathol ; 247(3): 320-332, 2019 03.
Article in English | MEDLINE | ID: mdl-30426505

ABSTRACT

Immune responses contribute to a large extent to heart diseases. However, it is still not clear how the key inflammatory mediator interferon-γ (IFNγ) plays a role in doxorubicin (DOX)-induced cardiomyopathy. We report here that DOX-induced heart dysfunction involves IFNγ signaling in mice. The IFNγ receptor was found to be highly expressed on cardiomyocytes, and its downstream signaling was activated in heart tissues upon DOX treatment. In vitro, IFNγ strongly aggravated the injury of cardiomyocytes exposed to DOX. Although not affecting DOX-induced cell death, IFNγ disrupted mitochondrial respiration and fatty acid oxidation in DOX-exposed cardiomyocytes. IFNγ extended the suppression of the AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) axis by DOX to a p38-dependent branch. Activation of AMPK or inhibition of p38 inhibited the enhancing effect of IFNγ on the DOX-induced cardiotoxicity and prolonged the survival time in DOX-treated mice. Taken together, our results indicate that reprogramming of cardiac metabolism by IFNγ represents a previously unidentified key step for DOX-induced cardiomyopathy. This unavoidable impact of IFNγ on cardiomyocyte metabolism during chemotherapy redirects our attention to the balance between beneficial immunosurveillance of cancer cells and unwanted toxic side-effects. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiotoxicity/immunology , Doxorubicin/toxicity , Interferon-gamma/immunology , Myocytes, Cardiac/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Cell Respiration/physiology , Cells, Cultured , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Doxorubicin/pharmacology , Fatty Acids/metabolism , Female , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocytes, Cardiac/immunology , Oxidation-Reduction , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
6.
Front Immunol ; 9: 1776, 2018.
Article in English | MEDLINE | ID: mdl-30127784

ABSTRACT

S100A4, a calcium-binding protein, can promote pulmonary fibrosis via fibroblast activation. Due partly to its various cellular origins, the exact role of S100A4 in the development of lung fibrosis remains elusive. Here, we show that in the bronchoalveolar lavage fluid, numbers of S100A4+ macrophages correlated well with S100A4 protein levels and occurrence of idiopathic pulmonary fibrosis (IPF) in patients. A mouse model of bleomycin-induced pulmonary fibrosis demonstrated S100A4+ macrophages as main source for extracellular S100A4 in the inflammatory phase. In vitro studies revealed that extracellular S100A4 could activate both mouse and human lung fibroblasts by upregulation of α-SMA and type I collagen, during which sphingosine-1-phosphate (S1P) increased. Inhibiting the S1P receptor subtypes S1P1/S1P3 abrogated fibroblast activation. Accordingly, absence or neutralization of S100A4 significantly attenuated bleomycin-induced lung fibrosis in vivo. Importantly, adoptive transfer of S100A4+ but not of S100A4- macrophages installed experimental lung injury in S100A4-/- mice that were otherwise not sensitive to fibrosis induction. Taken together, S100A4 released by macrophages promotes pulmonary fibrosis through activation of lung fibroblasts which is associated with S1P. This suggests that extracellular S100A4 or S100A4+ macrophages within the lung as promising targets for early clinical diagnosis or therapy of IPF.


Subject(s)
Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , S100 Calcium-Binding Protein A4/metabolism , Actins/metabolism , Animals , Bleomycin/pharmacology , Bronchoalveolar Lavage Fluid , CD11b Antigen , Cell Line , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
7.
Front Immunol ; 9: 388, 2018.
Article in English | MEDLINE | ID: mdl-29556233

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) often expand during cancer or chronic inflammation and dampen immune responses. However, mechanisms underlying their capacity to escape intrinsic apoptosis in the inflammatory environment are still largely unknown. In this study, we investigated this in mouse tumor models with MDSC accumulation. Spontaneous rejection of tumors implanted into mice deficient for the small Ca2+-binding protein S100A4 (S100A4-/-) was accompanied by low numbers of peripheral MDSCs. This was independent of S100A4 expression on tumor cells. In contrast, MDSCs from S100A4-/- tumor-bearing mice showed a diminished resistance to the induction of intrinsic apoptosis. Further studies demonstrated that S100A4 protects MDSCs from apoptosis through toll-like receptor-4/extracellular signal-regulated kinase-dependent caspase-9 inhibition. The finding that S100A4 is critical for MDSC survival in inflammatory environments might have important implications for the clinical treatment of cancer or inflammation-related diseases.


Subject(s)
Inflammation/metabolism , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/metabolism , S100 Calcium-Binding Protein A4/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Neoplasms, Experimental/immunology , S100 Calcium-Binding Protein A4/genetics , Toll-Like Receptor 4/genetics
8.
J Cancer Res Clin Oncol ; 144(3): 499-507, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29327244

ABSTRACT

PURPOSE: To analyze the impact of TNFα or IL2 on human lymphocytes in vitro and the anti-tumor and immune-modifying effects of L19-IL2 and L19-TNFα on subcutaneously growing J558L myeloma in immunocompetent mice. METHODS: PBMCs from three healthy volunteers were incubated with IL2, TNFα, or with IL2 plus addition of TNFα (final 20 h). BALB/c J558L mice with subcutaneous tumors were treated with intravenous L19-TNFα plus L19-IL2, or controls. Tumor growth and intra- and peri-tumoral tissues were analyzed for micro-vessel density, necrosis, immune cell composition, and PD1 or PD-L1 expressing cells. RESULTS: Exposure of PBMC in vitro to IL2, TNFα, or to IL2 over 3 and 5 days plus TNFα for the final 20 h resulted in an approximately 50 and 75% reduction of the CD25low effector cell/CD25high Treg cell ratio, respectively, compared to medium control. IL2 or TNFα increased the proportion of CD4- CD25low effector lymphocytes while reducing the proportion of CD4+ CD25low Teff cells. In the J558L myeloma model, tumor eradication was observed in 58, 42, 25, and 0% of mice treated with L19-TNFα plus L19-IL2, L19-TNFα, L19-IL2, and PBS, respectively. L19-TNFα/L19-IL2 combination caused tumor necrosis, capillary density doubling, peri-tumoral T cell and PD1+ T cell reduction (- 50%), and an increase in PD-L1+ myeloma cells. CONCLUSION: IL2, TNFα, or IL2 plus TNFα (final 20 h) increased the proportion of CD4- CD25low effector lymphocytes possibly indicating immune activation. L19-TNFα/L19-IL2 combination therapy eradicated tumors in J558L myeloma BALB/c mice likely via TNFα-induced tumor necrosis and L19-TNFα/L19-IL2-mediated local cellular immune reactions.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Interleukin-2/therapeutic use , Multiple Myeloma/therapy , Neovascularization, Pathologic/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Female , Immunotoxins/therapeutic use , Mice , Mice, Inbred BALB C , Multiple Myeloma/immunology , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transplantation, Isogeneic
9.
Hematol Oncol ; 36(1): 291-298, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28547857

ABSTRACT

Graft-versus-host disease (GvHD) is a major complication of allogeneic stem cell transplantation. High-resolution in vivo histology of the intestine by confocal endomicroscopy (CEM) detects acute GvHD (aGvHD) with high sensitivity. This pilot study aims to evaluate the diagnostic value of CEM for intestinal chronic GvHD (cGvHD). The study included 20 patients with gastrointestinal symptoms and confirmed cGvHD in other organs as well as 20 patients with clinically suspected acute GvHD for control. Confocal endomicroscopy was performed as gastroscopy followed by sigmoidoscopy after intravenous injection of fluorescein (10%) and topical application of acriflavine (0.05%). Histopathology from H&E-stained biopsy samples throughout the intestinal tract complemented the survey. All histological features of intestinal cGvHD were predominantly mild to moderate. Stroma fibrosis detected by standard histology (16/20 patients) was not seen by CEM. Apoptosis assessed by histology in 12/20 patients was concordant with CEM (8/12 patients). Confocal endomicroscopy revealed esophageal manifestation of cGvHD in 3 patients. For each biopsy site, CEM correlated with intestinal histology (r = 0.64). Classical histology from intestinal biopsy samples taken under CEM monitoring confirmed the final diagnosis of cGvHD. The sensitivity of CEM with 40% in cGvHD was significantly lower compared to 70% in patients with aGvHD. Confocal endomicroscopy detected acute features of cGvHD and contributed to the diagnosis of esophageal cGvHD but failed to display stroma fibrosis in vivo. Although CEM represents a useful noninvasive tool in routine diagnostic of intestinal aGvHD, the method is not sufficient to fully establish the diagnosis of cGvHD within the intestinal tract. Confocal endomicroscopy allowed acquisition of targeted biopsies in patients suspected of having cGvHD.


Subject(s)
Gastrointestinal Diseases/diagnostic imaging , Graft vs Host Disease/diagnostic imaging , Microscopy, Confocal/methods , Adult , Aged , Chronic Disease , Female , Gastrointestinal Diseases/pathology , Graft vs Host Disease/pathology , Humans , Male , Microscopy, Confocal/instrumentation , Middle Aged
10.
Infect Immun ; 85(8)2017 08.
Article in English | MEDLINE | ID: mdl-28559404

ABSTRACT

Classical Whipple's disease (CWD) is characterized by the lack of specific Th1 response toward Tropheryma whipplei in genetically predisposed individuals. The cofactor GrpE of heat shock protein 70 (Hsp70) from T. whipplei was previously identified as a B-cell antigen. We tested the capacity of Hsp70 and GrpE to elicit specific proinflammatory T-cell responses. Peripheral mononuclear cells from CWD patients and healthy donors were stimulated with T. whipplei lysate or recombinant GrpE or Hsp70 before levels of CD40L, CD69, perforin, granzyme B, CD107a, and gamma interferon (IFN-γ) were determined in T cells by flow cytometry. Upon stimulation with total bacterial lysate or recombinant GrpE or Hsp70 of T. whipplei, the proportions of activated effector CD4+ T cells, determined as CD40L+ IFN-γ+, were significantly lower in patients with CWD than in healthy controls; CD8+ T cells of untreated CWD patients revealed an enhanced activation toward unspecific stimulation and T. whipplei-specific degranulation, although CD69+ IFN-γ+ CD8+ T cells were reduced upon stimulation with T. whipplei lysate and recombinant T. whipplei-derived proteins. Hsp70 and its cofactor GrpE are immunogenic in healthy individuals, eliciting effective responses against T. whipplei to control bacterial spreading. The lack of specific T-cell responses against these T. whipplei-derived proteins may contribute to the pathogenesis of CWD.


Subject(s)
Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HSP70 Heat-Shock Proteins/immunology , Heat-Shock Proteins/immunology , Tropheryma/immunology , Whipple Disease/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , B-Lymphocytes/pathology , Duodenum/immunology , Female , Flow Cytometry , Humans , Interferon-gamma/genetics , Intestinal Mucosa/immunology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation , Male , Middle Aged , Tropheryma/chemistry , Tropheryma/genetics , Whipple Disease/physiopathology , Young Adult
11.
J Pathol ; 242(3): 334-346, 2017 07.
Article in English | MEDLINE | ID: mdl-28418194

ABSTRACT

Angiostasis mediated by interferon (IFN)-γ is a key mechanism of anti-tumour immunity; however, the effect of IFN-γ on host vascular endothelial growth factor A (VEGFA)-expressing cells during tumour progression is still elusive. Here, we developed transgenic mice with IFN-γ receptor (IFNγR) expression under control of the Vegfa promoter (V-γR). In these mice, the IFN-γ responsiveness of VEGFA-expressing cells led to dramatic growth suppression of transplanted lung carcinoma cells. Surprisingly, increased mortality and tumour metastasis were observed in the tumour-bearing V-γR mice, in comparison with the control wild-type and IFNγR-deficient mice. Further study showed that perivascular cells were VEGFA-expressing cells and potential IFN-γ targets. In vivo, tumour vascular perfusion and pericyte association with blood vessels were massively disrupted in V-γR mice. In vitro, IFN-γ inhibited transforming growth factor-ß signalling by upregulating SMAD7, and therefore downregulated N-cadherin expression in pericytes. Importantly, IFN-γ neutralization in vivo with a monoclonal antibody reduced tumour metastasis. Together, the results suggest that IFNγR-mediated dissociation of perivascular cells from blood vessels contributes to the acceleration of tumour metastasis. Thus, the inhibition of tumour growth via IFN-γ-induced angiostasis might also accelerate tumour metastasis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Lung Neoplasms/physiopathology , Receptors, Interferon/physiology , Animals , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation , Fibroblasts/metabolism , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Pericytes/metabolism , Receptors, Interferon/deficiency , Receptors, Interferon/metabolism , Smad7 Protein/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/metabolism , Interferon gamma Receptor
12.
Cell Mol Immunol ; 14(7): 597-606, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27133471

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are well known for their capacity to suppress antitumor T-cell responses, but their effects on B-cell function and antibody production remain unclear. Here, we found that MDSCs that accumulated around the germinal center in the spleen of tumor-bearing mice co-located with B cells. In the presence of MDSCs, the antibody reaction to a surrogate antigen was significantly enhanced in mice, especially the immunoglobulin (Ig)A subtype. Co-culture with MDSCs promoted both proliferation and differentiation of B cells into IgA-producing plasma cells in vitro. Interestingly, the cross talk between MDSCs and B cells required cell-cell contact. MDSCs from tumor necrosis factor receptor (TNFR) 2-/- mice, but not from TNFR1-/- mice, failed to promote B-cell responses. Further investigation suggested that interleukin-10 and transforming growth factor-ß1 were crucial for the MDSC-mediated promotion of IgA responses. These results demonstrate a novel mechanism of MDSC-mediated immune regulation during tumor growth.


Subject(s)
B-Lymphocytes/metabolism , Immunoglobulin A/biosynthesis , Myeloid-Derived Suppressor Cells/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Animals , Antibody Formation , Cell Proliferation , Interleukin-10/metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Spleen/metabolism , Transforming Growth Factor beta1/metabolism
13.
Curr Opin Gastroenterol ; 33(1): 53-58, 2017 01.
Article in English | MEDLINE | ID: mdl-27798440

ABSTRACT

PURPOSE OF REVIEW: The composition of activated adipose tissue with adipocytes secreting a broad spectrum of immune-modulatory adipokines next to adipose tissue-derived stromal cells and professional immune effector cells in the visceral fat creates a complex network of inflammatory processes shaping local immune responses in the adjacent inflamed intestinal mucosa. RECENT FINDINGS: In Crohn's disease a particular phenomenon called 'creeping fat' can be observed. Here the hyperplastic mesenteric fat tissue not only grows around inflamed small intestinal segments but also furthermore affects the regulation of the mucosal immune system. Diverticular disease is highly prevalent in the western world but the knowledge about its immunopathology remains incomplete. Interestingly, adipose tissue also frequently covers the basolateral site of inflamed diverticula, hence locally reflecting the phenomenon seen in Crohn's disease. SUMMARY: This review aims to summarize the current knowledge in which measures this intraabdominal fat participates in the regulation of intestinal inflammation with a particular focus on differences and possible parallels in Crohn's disease and diverticulitis. The available data allow for suggesting that each inflamed diverticula mechanistically reflects Crohn's disease on a miniature scale.


Subject(s)
Adipokines/immunology , Adipose Tissue/immunology , Crohn Disease/immunology , Diverticulitis, Colonic/immunology , Inflammation/immunology , Intra-Abdominal Fat/immunology , Adipokines/metabolism , Adipose Tissue/metabolism , Colon/immunology , Colon/pathology , Crohn Disease/pathology , Crohn Disease/physiopathology , Diverticulitis, Colonic/pathology , Diverticulitis, Colonic/physiopathology , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/physiopathology
14.
Cancer Lett ; 378(1): 51-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27149931

ABSTRACT

Carboxylesterase 2 (CES-2) is instrumental for conversion of ester-containing prodrugs in cancer treatment. CES-2 expression was analyzed by immunohistochemistry in colorectal cancer (CRC) compared to colonic inflammation as well as in liver and peripheral blood. In CRC, tumor grades showed no correlation with levels of CES-2 expression, which was heterogeneous within these tumors. Cellular infiltrates in the immediate tumor vicinity expressed high levels of CES-2. Thus, tissue adjacent to the tumor was a substantial source of CES-2 with high expression in plasma cells. CES-2(high) plasma cells were abundantly found in the colon of patients with inflammatory bowel disease. CES-2 expression is strong in hepatocytes of normal livers, while CES-2 expression in peripheral blood mononuclear cells of healthy donors was overall low at protein and mRNA levels. In summary, the conversion of ester-containing prodrugs by CES-2 is mainly to occur in the periphery, during liver passage and in the colon after enterohepatic recirculation. We here demonstrated plasma cells as strong producers of CES-2. Further studies should elucidate the role of CES-2(+) plasma cells in intestinal inflammation and cancer.


Subject(s)
Antineoplastic Agents/metabolism , Carboxylesterase/metabolism , Colorectal Neoplasms/enzymology , Gastrointestinal Agents/metabolism , Inflammatory Bowel Diseases/enzymology , Plasma Cells/enzymology , Prodrugs/metabolism , Activation, Metabolic , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carboxylesterase/blood , Carboxylesterase/genetics , Colon/enzymology , Colon/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Enterohepatic Circulation , Female , Gastrointestinal Agents/pharmacology , Gene Expression Regulation, Enzymologic , HEK293 Cells , HT29 Cells , Hepatocytes/enzymology , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Jurkat Cells , K562 Cells , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Neoplasm Grading , Prodrugs/therapeutic use , U937 Cells , Young Adult
15.
Inflamm Intest Dis ; 1(2): 52-62, 2016 Jul.
Article in English | MEDLINE | ID: mdl-29922658

ABSTRACT

BACKGROUND: Chronic intestinal inflammation due to noninfectious causes represents a growing health issue all over the world. Celiac disease as well as inflammatory bowel diseases (IBD) like Crohn's disease and ulcerative and microscopic colitis involve uncontrolled T-cell activation and T-cell-mediated damage as common denominators. Therefore, diagnosis and treatment decisions clearly benefit from the knowledge of the intricacies of the systemic and the local T-cell activity. SUMMARY: Depending on the cytokine milieu, CD4+ T cells can differentiate into proinflammatory T helper 1 (Th1), anti-inflammatory Th2, antimicrobial Th17, pleiotropic Th9, tissue-instructing Th22 cells, and in the regulatory compartment forkhead box protein 3+ Treg, suppressive Tr1 or Th3 cells. Additionally, follicular Th cells provide B-cell help in antibody class switching; cytotoxic CD8+ T cells target virus-infected or tumor cells. This review discusses our current knowledge on the contribution of defined T-cell subpopulations to establishing and maintaining chronic intestinal inflammation in either of the above entities. It also puts emphasis on the differences in the prevalence of these diseases between Eastern and Western countries. KEY MESSAGES: In celiac disease, the driving role of T cells in the lamina propria and in the epithelium mainly specific for two defined antigens is well established. Differences in genetics and lifestyle between Western and Eastern countries were instrumental in understanding underlying mechanisms. In IBD, the vast amount of potential antigens and the corresponding antigen-specific T cells makes it unlikely to find universal triggers. Increased mucosal CD4+ regulatory T cells in all four entities fail to control or abrogate local inflammatory processes. Thus, prevailing differences in the functional T-cell subtypes driving chronic intestinal inflammation in celiac disease and IBD at best allow some overlap in the treatment options for either disease.

16.
Front Immunol ; 6: 613, 2015.
Article in English | MEDLINE | ID: mdl-26697009

ABSTRACT

Macrophages as innate immune cells and fast responders to antigens play a central role in protecting the body from the luminal content at a huge interface. Chronic inflammation in inflammatory bowel diseases massively alters the number and the subset diversity of intestinal macrophages. We here address the diversity within the human intestinal macrophage compartment at the level of similarities and differences between homeostasis and chronic intestinal inflammation as well as between UC and CD, including the potential role of macrophage subsets for intestinal fibrosis. Hallmark of macrophages is their enormous plasticity, i.e., their capacity to integrate signals from their environment thereby changing their phenotype and functions. Tissue-resident macrophages located directly beneath the surface epithelium in gut homeostasis are mostly tolerogenic. The total number of macrophages increases with luminal contents entering the mucosa through a broken intestinal barrier in ulcerative colitis (UC) as well as in Crohn's disease (CD). Although not fully understood, the resulting mixtures of tissue-resident and tissue-infiltrating macrophages in both entities are diverse with respect to their phenotypes and their distribution. Macrophages in UC mainly act within the intestinal mucosa. In CD, macrophages can also be found in the muscularis and the mesenteric fat tissue compartment. Taken together, the present knowledge on human intestinal macrophages so far provides a good starting point to dig deeper into the similarities and differences of functional subsets and to finally use their phenotypical diversity as markers for complex local milieus in health and disease.

17.
BMC Cancer ; 15: 663, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26450156

ABSTRACT

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease with the need for treatment optimization. Previously, high expression of Insulin-like growth factor binding protein 7 (IGFBP7), a member of the IGF system, was identified as negative prognostic factor in adult T-ALL patients. Since aberrant IGFBP7 expression was observed in a variety of neoplasia and was relevant for prognosis in T-ALL, we investigated the functional role of IGFBP7 in Jurkat and Molt-4 cells as in vitro models for T-ALL. METHODS: Jurkat and Molt-4 cells were stably transfected with an IGFBP7 over-expression vector or the empty vector as control. Proliferation of the cells was assessed by WST-1 assays and cell cycle status was measured by flow-cytometry after BrDU/7-AAD staining. The effect of IGFBP7 over-expression on sensitivity to cytostatic drugs was determined in AnnexinV/7-AAD assays. IGF1-R protein expression was measured by Western Blot and flow-cytometric analysis. IGF1-R associated gene expression profiles were generated from microarray gene expression data of 86 T-ALL patients from the Microarrays Innovations in Leukemia (MILE) multicenter study. RESULTS: IGFBP7-transfected Jurkat cells proliferated less, leading to a longer survival in a nutrient-limited environment. Both IGFBP7-transfected Jurkat and Molt-4 cells showed an arrest in the G0/G1 cell cycle phase. Furthermore, Jurkat IGFBP7-transfected cells were resistant to vincristine and asparaginase treatment. Surface expression and whole protein measurement of IGF1-R protein expression showed a reduced abundance of the receptor after IGFBP7 transfection in Jurkat cells. Interestingly, combination of the IGF1-R inhibitor NPV-AEW541 restored sensitivity to vincristine in IGFBP7-transfected cells. Additionally, IGF1-R associated GEP revealed an up-regulation of important drivers of T-ALL pathogenesis and regulators of chemo-resistance and apoptosis such as NOTCH1, BCL-2, PRKCI, and TP53. CONCLUSION: This study revealed a proliferation inhibiting effect of IGFBP7 by G0/G1 arrest and a drug resistance-inducing effect of IGFBP7 against vincristine and asparaginase in T-ALL. These results provide a model for the previously observed association between high IGFBP7 expression and chemotherapy failure in T-ALL patients. Since the resistance against vincristine was abolished by IGF1-R inhibition, IGFBP7 could serve as biomarker for patients who may benefit from therapies including IGF1-R inhibitors in combination with chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Profiling , Humans , Jurkat Cells , Receptor, IGF Type 1 , Transcriptome
18.
J Cell Mol Med ; 19(9): 2162-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26059794

ABSTRACT

Modifying the chromatin structure and interacting with non-histone proteins, histone deacetylases (HDAC) are involved in vital cellular processes at different levels. We here specifically investigated the direct effects of HDAC5 in macrophage activation in response to bacterial or cytokine stimuli. Using murine and human macrophage cell lines, we studied the expression profile and the immunological function of HDAC5 at transcription and protein level in over-expression as well as RNA interference experiments. Toll-like receptor-mediated stimulation of murine RAW264.7 cells significantly reduced HDAC5 mRNA within 7 hrs but presented baseline levels after 24 hrs, a mechanism that was also found for Interferon-γ treatment. If treated with lipopolysaccharide, RAW264.7 cells transfected for over-expression only of full-length but not of mutant HDAC5, significantly elevated secretion of tumour necrosis factor α and of the monocyte chemotactic protein-1. These effects were accompanied by increased nuclear factor-κB activity. Accordingly, knock down of HDAC5-mRNA expression using specific siRNA significantly reduced the production of these cytokines in RAW264.7 or human U937 cells. Taken together, our results suggest a strong regulatory function of HDAC5 in the pro-inflammatory response of macrophages.


Subject(s)
Histone Deacetylases/metabolism , Inflammation/enzymology , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Animals , Cytokines/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Humans , Kinetics , Mice , NF-kappa B/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , U937 Cells
19.
PLoS One ; 10(6): e0123867, 2015.
Article in English | MEDLINE | ID: mdl-26052942

ABSTRACT

Leukocyte adhesion and transmigration are central features governing immune surveillance and inflammatory reactions in body tissues. Within the liver sinusoids, chemokines initiate the first crucial step of T-cell migration into the hepatic tissue. We studied molecular mechanisms involved in endothelial chemokine supply during hepatic immune surveillance and liver inflammation and their impact on the recruitment of CD4(+) T cells into the liver. In the murine model of Concanavalin A-induced T cell-mediated hepatitis, we showed that hepatic expression of the inflammatory CXC chemokine ligands (CXCL)9 and CXCL10 strongly increased whereas homeostatic CXCL12 significantly decreased. Consistently, CD4(+) T cells expressing the CXC chemokine receptor (CXCR)3 accumulated within the inflamed liver tissue. In histology, CXCL9 was associated with liver sinusoidal endothelial cells (LSEC) which represent the first contact site for T-cell immigration into the liver. LSEC actively transferred basolaterally internalized CXCL12, CXCL9 and CXCL10 via clathrin-coated vesicles to CD4(+) T cells leading to enhanced transmigration of CXCR4(+) total CD4(+) T cells and CXCR3(+) effector/memory CD4(+) T cells, respectively in vitro. LSEC-expressed CXCR4 mediated CXCL12 transport and blockage of endothelial CXCR4 inhibited CXCL12-dependent CD4(+) T-cell transmigration. In contrast, CXCR3 was not involved in the endothelial transport of its ligands CXCL9 and CXCL10. The clathrin-specific inhibitor chlorpromazine blocked endothelial chemokine internalization and CD4(+) T-cell transmigration in vitro as well as migration of CD4(+) T cells into the inflamed liver in vivo. Moreover, hepatic accumulation of CXCR3(+) CD4(+) T cells during T cell-mediated hepatitis was strongly reduced after administration of chlorpromazine. These data demonstrate that LSEC actively provide perivascularly expressed homeostatic and inflammatory chemokines by CXCR4- and clathrin-dependent intracellular transport mechanisms thereby contributing to the hepatic recruitment of CD4(+) T-cell populations during immune surveillance and liver inflammation.


Subject(s)
Chemokine CXCL10/metabolism , Chemokine CXCL12/metabolism , Chemokine CXCL9/metabolism , Endothelial Cells/metabolism , Liver/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , Caveolae/drug effects , Caveolae/metabolism , Chlorpromazine/pharmacology , Clathrin/metabolism , Clathrin-Coated Vesicles/drug effects , Clathrin-Coated Vesicles/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Endothelial Cells/drug effects , Hepatitis/immunology , Hepatitis/pathology , Homeostasis/drug effects , Inflammation/pathology , Inflammation Mediators/metabolism , Liver/drug effects , Liver/immunology , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL
20.
Inflamm Bowel Dis ; 21(6): 1297-305, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25901973

ABSTRACT

BACKGROUND: Macrophages are key players in inflammatory bowel diseases (IBD). This study aimed to determine site-specific effects of defined macrophage subtypes on the integrity of the intestinal epithelial barrier. METHODS: Macrophage subtypes in situ in intestinal specimens of patients with IBD were visualized by immunohistochemistry. In vitro polarization of human peripheral CD14 cells yielded M1 or M2 macrophages. The influence of primary monocytes or macrophage subtypes on epithelial barrier integrity was analyzed by transepithelial resistance measurements, Western blot analysis, confocal laser scanning microscopy, and cytometric bead array in a coculture model of primary human macrophages and layers of intestinal epithelial cell lines. RESULTS: The lamina propria of the inflamed intestine in patients with IBD, predominantly in Crohn's disease, is massively infiltrated by CD68 cells also positive for inducible nitric oxide synthase and tumor necrosis factor (TNF) α. The presence of M1 macrophage shifted the balance in the local macrophage compartment towards a proinflammatory state. In the coculture model, monocytes and M1 macrophages reduced transepithelial resistance as a marker for epithelial barrier integrity. The mechanisms for paracellular leakage included intracellular relocalization of tight junction proteins like claudin-2 and epithelial cell apoptosis. Determined by specific cytokine blockade, M1 macrophages exerted their deleterious effect mainly through TNF-α, whereas monocyte-mediated damage was driven by the inflammasome effector cytokines, interleukin-1ß and interleukin-18. CONCLUSIONS: Lamina propria monocytes and M1 macrophages invading intestinal tissues directly contribute to disrupting the epithelial barrier through deregulation of tight junction proteins and induction of epithelial cell apoptosis, thus driving intestinal inflammation in IBD.


Subject(s)
Inflammatory Bowel Diseases/etiology , Intestines/pathology , Macrophages/cytology , Monocytes/cytology , Adolescent , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis , Claudin-2/metabolism , Cytokines/metabolism , Epithelial Cells/physiology , Humans , Inflammasomes/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors , Macrophages/metabolism , Middle Aged , Monocytes/metabolism , Nitric Oxide Synthase/metabolism , Transendothelial and Transepithelial Migration/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...