Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 134: 51-61, 2019 09.
Article in English | MEDLINE | ID: mdl-31271758

ABSTRACT

Reduced shear stress resulting from disturbed blood flow can impair endothelial integrity and drive the development of vascular inflammatory lesions. Metalloproteinases of the ADAM family have been implicated in the regulation of cell survival and inflammatory responses. Here we investigate the mechanism and function of ADAM15 upregulation in primary flow cultured endothelial cells. Transcriptomic analysis indicated that within the ADAM family ADAM15 mRNA is most prominently upregulated (4-fold) when endothelial cells are exposed to physiologic shear stress. This induction was confirmed in venous, arterial and microvascular endothelial cells and is associated with increased presence of ADAM15 protein in the cell lysates (5.6-fold) and on the surface (3.1-fold). The ADAM15 promoter contains several consensus sites for the transcription factor KLF2 which is also upregulated by shear stress. Induction of endothelial KLF2 by simvastatin treatment is associated with ADAM15 upregulation (1.8-fold) which is suppressed by counteracting simvastatin with geranylgeranyl pyrophosphate. KLF2 overexpression promotes ADAM15 expression (2.1-fold) under static conditions whereas KLF2 siRNA knockdown prevents ADAM15 induction by shear stress. Functionally, ADAM15 promotes survival of endothelial cells challenged by growth factor depletion or TNF stimulation as shown by ADAM15 shRNA knockdown (1.6-fold). Exposure to shear stress increases endothelial survival while additional knockdown of ADAM15 reduces survival (6.7-fold) under flow conditions. Thus, physiologic shear stress resulting from laminar flow promotes KLF2 induced ADAM15 expression which contributes to endothelial survival. The absence of ADAM15 at low shear stress or static conditions may therefore lead to increased endothelial damage and promote vascular inflammation.


Subject(s)
ADAM Proteins/genetics , Endothelial Cells/physiology , Membrane Proteins/genetics , Up-Regulation/genetics , Cells, Cultured , Endothelium, Vascular/physiology , Gene Expression Regulation/genetics , Human Umbilical Vein Endothelial Cells , Humans , RNA, Messenger/genetics , Stress, Mechanical
2.
Atherosclerosis ; 286: 163-171, 2019 07.
Article in English | MEDLINE | ID: mdl-30910225

ABSTRACT

BACKGROUND AND AIMS: Members of the family of a disintegrin and metalloproteinases (ADAMs) and their substrates have been previously shown to modulate the inflammatory response in cardiac diseases, but studies investigating the relevance of ADAM8 are still rare. Our aim is to provide evidence for the inflammatory dysregulation of ADAM8 in vascular diseases and its association with disease severity. METHODS: Western-type diet fed Apoe-/- and Ldlr-/- mice and artery ligation served as murine model for atherosclerosis and myocardial infarction, respectively. Human bypass grafts were used to study the association with coronary artery disease (CAD), with the simplified acute physiology score II (SAPS II) as a measure of postoperative organ dysfunction. Human primary vascular and blood cells were analyzed under basal and inflammatory conditions. mRNA levels were determined by RT-qPCR, ADAM8 protein levels by ELISA, immunohistochemistry or flow cytometry. RESULTS: ADAM8/ADAM8 expression is associated with atherosclerosis and CAD such as myocardial infarction in both mice and humans, especially in endothelial cells and leukocytes. We observed a strong in vivo and in vitro correlation of ADAM8 with the vascular disease markers VCAM-1, ICAM-1, TNF, IL-6, and CCL-2. Serum analysis revealed a significant elevation of soluble ADAM8 serum levels correlating with soluble CXCL16 levels and SAPS II. CONCLUSIONS: We demonstrate a general association of ADAM8 with cardiovascular diseases in mice and humans predominantly acting in endothelial cells and leukocytes. The correlation with postoperative organ dysfunctions in CAD patients highlights the value of further studies investigating the specific function of ADAM8 in cardiovascular diseases.


Subject(s)
ADAM Proteins/biosynthesis , Antigens, CD/biosynthesis , Atherosclerosis/metabolism , Membrane Proteins/biosynthesis , Myocardial Infarction/metabolism , Animals , Cells, Cultured , Female , Humans , Leukocytes, Mononuclear , Male , Mice , Middle Aged , Severity of Illness Index
3.
J Steroid Biochem Mol Biol ; 178: 340-347, 2018 04.
Article in English | MEDLINE | ID: mdl-29448043

ABSTRACT

Estrogens modulate the immune system and possess anti-inflammatory properties. In line, immune cells express a variety of estrogen receptors (ER) including ER-alpha and -beta. In the present study, we examined the influence of 17beta-estradiol (E2) serum concentrations on blood leukocyte composition and their ex vivo polarization/activation status by FACS analysis in sub-fertile human females under controlled ovarian stimulation (COS). Using a set of cell-type and polarization-specific markers, we demonstrate that increased 17ß-estradiol (E2) serum concentrations yield an overall increase in leukocytes, neutrophils and monocytes but decreased lymphocytes. There was a clear ratio shift towards an increase in M2 monocytes with a protective quality and an increase in T-helper cells compared to a decrease in cytotoxic T-cells. These data support experimental findings and clinical trials, i.e. related to multiple sclerosis and other autoimmune-related diseases, that have shown a down-regulation of CD8(+) T cells and up-regulation of T-regulatory cells. Further studies have to pinpoint to which extent the immune system/-responsiveness of otherwise healthy female patients is affected by medium-term systemic E2 variations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Estrogens/blood , Infertility, Female/immunology , Leukocytes/immunology , Neutrophils/immunology , Ovulation Induction , Receptors, Estrogen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Humans , Infertility, Female/blood , Infertility, Female/pathology , Leukocytes/metabolism , Neutrophils/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
4.
Glia ; 65(12): 1900-1913, 2017 12.
Article in English | MEDLINE | ID: mdl-28836302

ABSTRACT

Brain-intrinsic degenerative cascades are a proposed factor driving inflammatory lesion formation in multiple sclerosis (MS) patients. We recently described a model combining noninflammatory cytodegeneration (via cuprizone) with the classic active experimental autoimmune encephalomyelitis (Cup/EAE model), which exhibits inflammatory forebrain lesions. Here, we describe the histopathological characteristics and progression of these Cup/EAE lesions. We show that inflammatory lesions develop at various topographical sites in the forebrain, including white matter tracts and cortical and subcortical grey matter areas. The lesions are characterized by focal demyelination, discontinuation of the perivascular glia limitans, focal axonal damage, and neutrophil granulocyte extravasation. Transgenic mice with enhanced green fluorescent protein-expressing microglia and red fluorescent protein-expressing monocytes reveal that both myeloid cell populations contribute to forebrain inflammatory infiltrates. EAE-triggered inflammatory cerebellar lesions were augmented in mice pre-intoxicated with cuprizone. Gene expression studies suggest roles of the chemokines Cxcl10, Ccl2, and Ccl3 in inflammatory lesion formation. Finally, follow-up experiments in Cup/EAE mice with chronic disease revealed that forebrain, but not spinal cord, lesions undergo spontaneous reorganization and repair. This study underpins the significance of brain-intrinsic degenerative cascades for immune cell recruitment and, in consequence, MS lesion formation.


Subject(s)
Disease Progression , Encephalitis/etiology , Encephalitis/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Sesquiterpenes/toxicity , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Encephalitis/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Freund's Adjuvant/toxicity , Gene Expression/drug effects , Gene Expression/genetics , Glial Fibrillary Acidic Protein/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Microglia/ultrastructure , Monocytes/pathology , Monocytes/ultrastructure , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L602-L614, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28596294

ABSTRACT

Alveolar leukocyte recruitment is a hallmark of acute lung inflammation and involves transmigration of leukocytes through endothelial and epithelial layers. The disintegrin and metalloproteinase (ADAM) 8 is expressed on human isolated leukocytic cells and can be further upregulated on cultured endothelial and epithelial cells by proinflammatory cytokines. By shRNA-mediated knockdown we show that leukocytic ADAM8 is required on monocytic THP-1 cells for chemokine-induced chemotaxis as well as transendothelial and transepithelial migration. Furthermore, ADAM8 promotes αL-integrin upregulation and THP-1 cell adhesion to endothelial cells. On endothelial cells ADAM8 enhances transendothelial migration and increases cytokine-induced permeability. On epithelial cells the protease facilitates migration in a wound closure assay but does not affect transepithelial leukocyte migration. Blood leukocytes and bone marrow-derived macrophages (BMDM) from ADAM8-deficient mice show suppressed chemotactic response. Intranasal application of LPS to mice is accompanied with ADAM8 upregulation in the lung. In this model of acute lung inflammation ADAM8-deficient mice are protected against leukocyte infiltration. Finally, transfer experiments of BMDM in mice indicate that ADAM8 exerts a promigratory function predominantly on leukocytes. Our study provides in vitro and in vivo evidence that ADAM8 on leukocytes holds a proinflammatory function in acute lung inflammation by promoting alveolar leukocyte recruitment.


Subject(s)
ADAM Proteins/metabolism , Antigens, CD/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Membrane Proteins/metabolism , Pneumonia/metabolism , Pneumonia/pathology , ADAM Proteins/deficiency , ADAM Proteins/genetics , Acute Disease , Animals , Antigens, CD/genetics , Cell Adhesion , Cell Membrane Permeability , Chemotaxis , Cytokines/metabolism , Edema/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , HEK293 Cells , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wound Healing
6.
Mediators Inflamm ; 2017: 1515389, 2017.
Article in English | MEDLINE | ID: mdl-28522896

ABSTRACT

Flow conditions critically regulate endothelial cell functions in the vasculature. Reduced shear stress resulting from disturbed blood flow can drive the development of vascular inflammatory lesions. On endothelial cells, the transmembrane chemokine CX3CL1/fractalkine promotes vascular inflammation by functioning as a surface-expressed adhesion molecule and by becoming released as soluble chemoattractant for monocytic cells expressing the receptor CX3CR1. Here, we report that endothelial cells from human artery, vein, or microvasculature constitutively express CX3CL1 when cultured under static conditions. Stimulation with TNFα under static or very low shear stress conditions strongly upregulates CX3CL1 expression. By contrast, CX3CL1 induction is profoundly reduced when cells are exposed to higher shear stress. When endothelial cells were grown and subsequently stimulated with TNFα under low shear stress, strong adhesion of monocytic THP-1 cells to endothelial cells was observed. This adhesion was in part mediated by transmembrane CX3CL1 as demonstrated with a neutralizing antibody. By contrast, no CX3CL1-dependent adhesion to stimulated endothelium was observed at high shear stress. Thus, during early stages of vascular inflammation, low shear stress typically seen at atherosclerosis-prone regions promotes the induction of endothelial CX3CL1 and monocytic cell recruitment, whereas physiological shear stress counteracts this inflammatory activation of endothelial cells.


Subject(s)
Cell Adhesion/physiology , Chemokine CX3CL1/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Adhesion/genetics , Chemokine CX3CL1/genetics , Chemokine CXCL16/genetics , Chemokine CXCL16/metabolism , Endothelial Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Shear Strength/physiology , Stress, Mechanical , THP-1 Cells
7.
PLoS One ; 12(3): e0173486, 2017.
Article in English | MEDLINE | ID: mdl-28267793

ABSTRACT

The CXC-chemokine receptor 6 (CXCR6) is a class A GTP-binding protein-coupled receptor (GPCRs) that mediates adhesion of leukocytes by interacting with the transmembrane cell surface-expressed chemokine ligand 16 (CXCL16), and also regulates leukocyte migration by interacting with the soluble shed variant of CXCL16. In contrast to virtually all other chemokine receptors with chemotactic activity, CXCR6 carries a DRF motif instead of the typical DRY motif as a key element in receptor activation and G protein coupling. In this work, modeling analyses revealed that the phenylalanine F3.51 in CXCR6 might have impact on intramolecular interactions including hydrogen bonds by this possibly changing receptor function. Initial investigations with embryonic kidney HEK293 cells and further studies with monocytic THP-1 cells showed that mutation of DRF into DRY does not influence ligand binding, receptor internalization, receptor recycling, and protein kinase B (AKT) signaling. Adhesion was slightly decreased in a time-dependent manner. However, CXCL16-induced calcium signaling and migration were increased. Vice versa, when the DRY motif of the related receptor CX3CR1 was mutated into DRF the migratory response towards CX3CL1 was diminished, indicating that the presence of a DRF motif generally impairs chemotaxis in chemokine receptors. Transmembrane and soluble CXCL16 play divergent roles in homeostasis, inflammation, and cancer, which can be beneficial or detrimental. Therefore, the DRF motif of CXCR6 may display a receptor adaptation allowing adhesion and cell retention by transmembrane CXCL16 but reducing the chemotactic response to soluble CXCL16. This adaptation may avoid permanent or uncontrolled recruitment of inflammatory cells as well as cancer metastasis.


Subject(s)
Adaptation, Biological , Amino Acid Motifs , Cell Adhesion , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Amino Acid Sequence , Calcium Signaling , Cell Line , Cell Membrane , Cell Movement , Chemotaxis , Gene Expression , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR6 , Receptors, Chemokine/genetics , Receptors, Virus/genetics , Signal Transduction
8.
Platelets ; 28(4): 354-361, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27460023

ABSTRACT

Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.


Subject(s)
ADAM Proteins/metabolism , Inflammation/genetics , Membrane Proteins/metabolism , Metalloproteases/genetics , Humans
9.
Biochim Biophys Acta ; 1863(11): 2795-2808, 2016 11.
Article in English | MEDLINE | ID: mdl-27599715

ABSTRACT

By mediating proteolytic shedding on the cell surface the disintegrin and metalloproteinases ADAM10 and ADAM17 function as critical regulators of growth factors, cytokines and adhesion molecules. We here report that stimulation of lung epithelial A549 tumor cells with phorbol-12-myristate-13-acetate (PMA) leads to the downregulation of the surface expressed mature form of ADAM17 without affecting ADAM10 expression. This reduction could not be sufficiently explained by metalloproteinase-mediated degradation, dynamin-mediated internalization or microdomain redistribution of ADAM17. Instead, surface downregulation of ADAM17 was correlated with the presence of its mature form in exosomes. Exosomal ADAM17 release was also observed in monocytic and primary endothelial cells where it could be induced by stimulation with lipopolysaccharide. Antibody-mediated surface labelling of ADAM17 revealed that at least part of exosomal ADAM17 was oriented with the metalloproteinase domain outside and had been expressed on the cell surface. Suppression of iRHOM2-mediated ADAM17 maturation prevented surface expression and exosomal release of ADAM17. Further, deletion of the protease's C-terminus or cell treatment with a calcium chelator diminished exosomal release as well as surface downregulation of ADAM17, underlining that both processes are closely associated. Co-incubation of ADAM17 containing exosomes with cells expressing the ADAM17 substrates TGFα or amphiregulin lead to increased shedding of both substrates. This was prevented when exosomes were prepared from cells with shRNA-mediated ADAM17 knockdown. These data indicate that cell stimulation can downregulate expression of mature ADAM17 from the cell surface and induce release of exosomal ADAM17, which can then distribute and contribute to substrate shedding on more distant cells.


Subject(s)
ADAM17 Protein/metabolism , Exosomes/enzymology , A549 Cells , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , Amphiregulin/metabolism , Amyloid Precursor Protein Secretases/metabolism , Calcium Signaling , Carrier Proteins/metabolism , Endothelial Cells/enzymology , Enzyme Activation , Exosomes/drug effects , Exosomes/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Membrane Microdomains/enzymology , Membrane Proteins/metabolism , Monocytes/enzymology , Protein Transport , RNA Interference , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Transforming Growth Factor alpha/metabolism
10.
Elife ; 52016 Jan 23.
Article in English | MEDLINE | ID: mdl-26802628

ABSTRACT

Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Axons/enzymology , Membrane Proteins/metabolism , Synapses/enzymology , ADAM10 Protein/deficiency , Amyloid Precursor Protein Secretases/deficiency , Animals , Axons/chemistry , Brain/embryology , Brain/enzymology , Gene Knockdown Techniques , Gene Knockout Techniques , Membrane Proteins/deficiency , Mice , Proteome/analysis , Substrate Specificity
11.
J Cell Mol Med ; 20(1): 104-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26499307

ABSTRACT

The chemokine CXCL16 and its receptor CXCR6 have been linked to the pathogenesis of acute and chronic cardiovascular disease. However, data on the clinical significance of CXCL16 in patients undergoing cardiac surgery with acute myocardial ischemia/reperfusion (I/R) are still lacking. Therefore, we determined CXCL16 in the serum of cardiac surgery patients and investigated its kinetics and association with the extent of organ dysfunction. 48 patients underwent conventional cardiac surgery with myocardial I/R and the use of cardiopulmonary bypass (CPB) were consecutively enrolled in the present study. We investigated the peri- and post-operative profile of CXCL16. Clinical relevant data were assessed and documented throughout the entire observation period. To identify the influence of myocardial I/R and CPB on CXCL16 release data were compared to those received from patients that underwent off-pump procedure. Pre-operative serum CXCL16 levels were comparable to those obtained from healthy volunteers (1174 ± 55.64 pg/ml versus 1225 ± 70.94). However, CXCL16 levels significantly increased during surgery (1174 ± 55.64 versus 1442 ± 75.42 pg/ml; P = 0.0057) and reached maximum levels 6 hrs after termination of surgery (1174 ± 55.64 versus 1648 ± 74.71 pg/ml; P < 0.001). We revealed a positive correlation between the intraoperative serum levels of CXCL16 and the extent of organ dysfunction (r(2) = 0.356; P = 0.031). Patients with high CXCL16 release showed an increased extent of organ dysfunction compared to patients with low CXCL16 release. Our study shows that CXCL16 is released into the circulation as a result of cardiac surgery and that high post-operative CXCL16 levels are associated with an increased severity of post-operative organ dysfunctions.


Subject(s)
Chemokines, CXC/blood , Heart Diseases/blood , Receptors, Scavenger/blood , Aged , Cells, Cultured , Chemokine CXCL16 , Female , Heart Diseases/immunology , Heart Diseases/surgery , Humans , Leukocyte Count , Male , Perioperative Period , Postoperative Complications/blood , Systemic Inflammatory Response Syndrome/blood , Treatment Outcome
12.
Oncotarget ; 6(31): 31295-312, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26378057

ABSTRACT

Syndecan-1 is a surface expressed heparan sulphate proteoglycan, which is upregulated by several tumor types and involved in tumor cell migration and metastasis. Syndecan-1 is shed from the cell surface and the remaining transmembrane fragment undergoes intramembrane proteolysis by γ-secretase. We here show that this generates a cytoplasmic C-terminal fragment (cCTF). In epithelial lung tumor A549 cells the endogenously produced cCTF accumulated when its proteasomal degradation was blocked with bortezomib and this accumulation was prevented by γ-secretase inhibition. Overexpression of the cCTF suppressed migration and invasion of A549 cells. This inhibitory effect was only seen when endogenous Syndecan-1 was present, but not in Syndecan-1 deficient cells. Further, overexpression of Syndecan-1 cCTF increased the basal activation of Src kinase, focal adhesion kinase (FAK) and Rho GTPase. This was associated with increased adhesion to fibronectin and collagen G and an increased recruitment of paxillin to focal adhesions. Moreover, lung tumor formation of A549 cells in mice was reduced by overexpression of Syndecan-1 cCTF. Finally, delivery of a synthetic peptide corresponding to the Syndecan-1 cCTF suppressed A549 cell migration and increased basal phosphorylation of Src and FAK. Our data indicate that the Syndecan-1 cCTF antagonizes Syndecan-1 dependent tumor cell migration in vitro and in vivo by dysregulating proadhesive signaling pathways and suggest that the cCTF can be used as an inhibitory peptide.


Subject(s)
Cell Movement , Cell Proliferation , Cytoplasm/metabolism , Lung Neoplasms/prevention & control , Peptide Fragments/pharmacology , Proteolysis , Syndecan-1/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, SCID , Phosphorylation , Signal Transduction , Syndecan-1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Hepatology ; 62(5): 1405-16, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26178698

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease is seen as the hepatic manifestation of the metabolic syndrome and represents the most common liver disease in Western societies. The G protein-coupled chemokine receptor CX3CR1 plays a central role in several metabolic syndrome-related disease manifestations and is involved in maintaining intestinal homeostasis. Because diet-induced intestinal dysbiosis is a driver for nonalcoholic fatty liver disease, we hypothesized that CX3CR1 may influence the development of steatohepatitis. In two independent models of diet-induced steatohepatitis (high-fat diet and methionine/choline-deficient diet), CX3CR1 protected mice from excessive hepatic steatosis and inflammation, as well as systemic glucose intolerance. Lack of Cx3cr1 expression was associated with significantly altered intestinal microbiota composition, which was linked to an impaired intestinal barrier. Concomitantly, endotoxin levels in portal serum and inflammatory macrophages in liver were increased in Cx3cr1-/- mice, indicating an increased inflammatory response. Depletion of intestinal microbiota by administration of broad-spectrum antibiotics suppressed the number of infiltrating macrophages and promoted macrophage polarization in liver. Consequently, antibiotic-treated mice demonstrated a marked improvement of steatohepatitis. CONCLUSION: Microbiota-mediated activation of the innate immune responses through CX3CR1 is crucial for controlling steatohepatitis progression, which recognizes CX3CR1 as an essential gatekeeper in this scenario.


Subject(s)
Homeostasis , Intestinal Mucosa/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Receptors, Chemokine/physiology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Translocation , Blood Glucose/analysis , CX3C Chemokine Receptor 1 , Immunity, Innate , Intestines/microbiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota , Non-alcoholic Fatty Liver Disease/metabolism
14.
Cell Mol Life Sci ; 72(19): 3783-801, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25912030

ABSTRACT

Syndecan-1 is a heparan sulfate proteoglycan expressed by endothelial and epithelial cells and involved in wound healing and tumor growth. Surface-expressed syndecan-1 undergoes proteolytic shedding leading to the release of the soluble N-terminal ectodomain from a transmembrane C-terminal fragment (tCTF). We show that the disintegrin and metalloproteinase (ADAM) 17 generates a syndecan-1 tCTF, which can then undergo further intra-membrane proteolysis by γ-secretase. Scratch-induced wound closure of cultured lung epithelial A549 tumor cells associates with increased syndecan-1 cleavage as evidenced by the release of shed syndecan-1 ectodomain and enhanced generation of the tCTF. Both wound closure and the associated syndecan-1 shedding can be suppressed by inhibition of ADAM family proteases. Cell proliferation, migration and invasion into matrigel as well as several signaling pathways implicated in these responses are suppressed by silencing of syndecan-1. These defects of syndecan-1 deficient cells can be overcome by overexpression of syndecan-1 tCTF or a corresponding tCTF of syndecan-4 but not by overexpression of a tCTF lacking the transmembrane domain. Finally, lung metastasis formation of A549 cells in SCID mice was found to be dependent on syndecan-1, and the presence of syndecan-1 tCTF was sufficient for this activity. Thus, the syndecan-1 tCTF by itself is capable of mediating critical syndecan-1-dependent functions in cell proliferation, migration, invasion and metastasis formation and therefore can replace full length syndecan-1 in the situation of increased syndecan-1 shedding during cell migration and tumor formation.


Subject(s)
ADAM Proteins/metabolism , Cell Movement/physiology , Epithelial Cells/physiology , Lung Neoplasms/secondary , Lung/cytology , Signal Transduction/physiology , Syndecan-1/metabolism , ADAM17 Protein , Animals , Blotting, Western , DNA Primers/genetics , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Mice , Mice, SCID , Polymerase Chain Reaction , Statistics, Nonparametric , Syndecan-1/chemistry
15.
Am J Pathol ; 185(4): 1145-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25659879

ABSTRACT

A disintegrin and metalloproteinase domain 10 (ADAM10) is a metalloprotease involved in cleavage of various cell surface molecules, such as adhesion molecules, chemokines, and growth factor receptors. Although we have previously shown an association of ADAM10 expression with atherosclerotic plaque progression, a causal role of ADAM10 in atherosclerosis has not been investigated. Bone marrow from conditional knockout mice lacking Adam10 in the myeloid lineage or from littermate controls was transplanted into lethally irradiated low density lipoprotein receptor Ldlr(-/-) mice on an atherogenic diet. Myeloid Adam10 deficiency did not affect plaque size, but it increased plaque collagen content. Matrix metalloproteinase 9 and 13 expression and matrix metalloproteinase 2 gelatinase activity were significantly impaired in Adam10-deficient macrophages, whereas their capacity to stimulate collagen production was unchanged. Furthermore, relative macrophage content in advanced atherosclerotic lesions was decreased. In vitro, Adam10-deficient macrophages showed reduced migration toward monocyte chemoattractant protein-1 and transmigration through collagen. In addition, Adam10-deficient macrophages displayed increased anti-inflammatory phenotype with elevated IL-10, and reduced production of proinflammatory tumor necrosis factor, IL-12, and nitric oxide in response to lipopolysaccharide. These data suggest a critical role of Adam10 for leukocyte recruitment, inflammatory mediator production, and extracellular matrix degradation. Thereby, myeloid ADAM10 may play a causal role in modulating atherosclerotic plaque stability.


Subject(s)
ADAM Proteins/deficiency , ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Inflammation/pathology , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , ADAM10 Protein , Animals , Collagen/metabolism , Cytokines/biosynthesis , Fibrosis , Flow Cytometry , Inflammation Mediators/metabolism , Integrases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Polymerase Chain Reaction , Receptors, LDL/deficiency , Receptors, LDL/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 308(4): L325-43, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25480335

ABSTRACT

Acute and chronic lung inflammation is driven and controlled by several endogenous mediators that undergo proteolytic conversion from surface-expressed proteins to soluble variants by a disintegrin and metalloproteinase (ADAM)-family members. TNF and epidermal growth factor receptor ligands are just some of the many substrates by which these proteases regulate inflammatory or regenerative processes in the lung. ADAM10 and ADAM17 are the most prominent members of this protease family. They are constitutively expressed in most lung cells and, as recent research has shown, are the pivotal shedding enzymes mediating acute lung inflammation in a cell-specific manner. ADAM17 promotes endothelial and epithelial permeability, transendothelial leukocyte migration, and inflammatory mediator production by smooth muscle and epithelial cells. ADAM10 is critical for leukocyte migration and alveolar leukocyte recruitment. ADAM10 also promotes allergic asthma by driving B cell responses. Additionally, ADAM10 acts as a receptor for Staphylococcus aureus (S. aureus) α-toxin and is crucial for bacterial virulence. ADAM8, ADAM9, ADAM15, and ADAM33 are upregulated during acute or chronic lung inflammation, and recent functional or genetic analyses have linked them to disease development. Pharmacological inhibitors that allow us to locally or systemically target and differentiate ADAM-family members in the lung suppress acute and asthmatic inflammatory responses and S. aureus virulence. These promising results encourage further research to develop therapeutic strategies based on selected ADAMs. These studies need also to address the role of the ADAMs in repair and regeneration in the lung to identify further therapeutic opportunities and possible side effects.


Subject(s)
ADAM Proteins/metabolism , Bacterial Toxins/metabolism , Gene Expression Regulation, Enzymologic , Hemolysin Proteins/metabolism , Pneumonia, Staphylococcal/metabolism , Proteolysis , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Animals , Asthma/metabolism , Asthma/pathology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Epidermal Growth Factor/metabolism , Humans , Pneumonia, Staphylococcal/pathology , Staphylococcal Infections , Transendothelial and Transepithelial Migration , Tumor Necrosis Factor-alpha/metabolism
17.
Blood ; 123(26): 4077-88, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24833351

ABSTRACT

Inflammation is a key process in various diseases, characterized by leukocyte recruitment to the inflammatory site. This study investigates the role of a disintegrin and a metalloproteinase (ADAM) 10 and ADAM17 for leukocyte migration in vitro and in a murine model of acute pulmonary inflammation. Inhibition experiments or RNA knockdown indicated that monocytic THP-1 cells and primary human neutrophils require ADAM10 but not ADAM17 for efficient chemokine-induced cell migration. Signaling and adhesion events that are linked to cell migration such as p38 and ρ GTPase-family activation, F-actin polymerization, adhesion to fibronectin, and up-regulation of α5 integrin were also dependent on ADAM10 but not ADAM17. This was confirmed with leukocytes isolated from mice lacking either ADAM10 or ADAM17 in all hematopoietic cells (vav 1 guanine nucleotide exchange factor [Vav]-Adam10(-/-) or Vav-Adam17(-/-) mice). In lipopolysaccharide-induced acute pulmonary inflammation, alveolar recruitment of neutrophils and monocytes was transiently increased in Vav-Adam17(-/-) but steadily reduced in Vav-Adam10(-/-) mice. This deficit in alveolar leukocyte recruitment was also observed in LysM-Adam10(-/-) mice lacking ADAM10 in myeloid cells and correlated with protection against edema formation. Thus, with regard to leukocyte migration, leukocyte-expressed ADAM10 but not ADAM17 displays proinflammatory activities and may therefore serve as a target to limit inflammatory cell recruitment.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Cell Movement , Membrane Proteins/metabolism , Neutrophil Infiltration , Neutrophils/enzymology , Pneumonia/enzymology , Pulmonary Alveoli/enzymology , Pulmonary Edema/enzymology , ADAM Proteins/genetics , ADAM10 Protein , ADAM17 Protein , Acute Disease , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/toxicity , Membrane Proteins/genetics , Mice , Mice, Knockout , Neutrophils/pathology , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , Pulmonary Alveoli/pathology , Pulmonary Edema/chemically induced , Pulmonary Edema/genetics , Pulmonary Edema/pathology
18.
J Immunol ; 192(2): 722-31, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24342803

ABSTRACT

In acute pulmonary inflammation, danger is first recognized by epithelial cells lining the alveolar lumen and relayed to vascular responses, including leukocyte recruitment and increased endothelial permeability. We supposed that this inflammatory relay critically depends on the immunological function of lung interstitial cells such as smooth muscle cells (SMC). Mice with smooth muscle protein-22α promotor-driven deficiency of the disintegrin and metalloproteinase (ADAM) 17 (SM22-Adam17(-/-)) were investigated in models of acute pulmonary inflammation (LPS, cytokine, and acid instillation). Underlying signaling mechanisms were identified in cultured tracheal SMC and verified by in vivo reconstitution experiments. SM22-Adam17(-/-) mice showed considerably decreased cytokine production and vascular responses in LPS- or acid-induced pulmonary inflammation. In vitro, ADAM17 deficiency abrogated cytokine release of primary SMC stimulated with LPS or supernatant of acid-exposed epithelial cells. This was explained by a loss of ADAM17-mediated growth factor shedding. LPS responses required ErbB1/epidermal growth factor receptor transactivation by TGFα, whereas acid responses required ErbB4 transactivation by neuregulins. Finally, LPS-induced pulmonary inflammation in SM22-Adam17(-/-) mice was restored by exogenous TGFα application, confirming the involvement of transactivation pathways in vivo. This highlights a new decisive immunological role of lung interstitial cells such as SMC in promoting acute pulmonary inflammation by ADAM17-dependent transactivation.


Subject(s)
ADAM Proteins/metabolism , ErbB Receptors/metabolism , Myocytes, Smooth Muscle/metabolism , Oncogene Proteins v-erbB/metabolism , Pneumonia/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/genetics , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Oncogene Proteins v-erbB/genetics , Pneumonia/genetics , Promoter Regions, Genetic/genetics , Receptor, ErbB-4 , Transcriptional Activation/genetics , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
19.
J Steroid Biochem Mol Biol ; 138: 195-205, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23792783

ABSTRACT

17ß-estradiol (E2) and progesterone (P) are neuroprotective hormones in different neurological disorders and in particular under hypoxic conditions in the brain. Both hormones dampen brain-intrinsic immune responses and regulate local glial cell function. Besides astrocytes which are functionally regulated in a manifold and complex manner, especially microglial cells are in the focus of steroid-mediated neuroprotection. In previous studies using a transient brain artery occlusion model, we demonstrated that microglial characteristics are critically modified after the administration of either E2 or P. We here studied the influence of sex steroids on the murine BV-2 microglia cell line under hypoxic conditions. Hypoxia changed the cell morphology from an amoeboid-like phenotype with processes to a rounded shape of secreting cell type. BV-2 cells expressed both estrogen receptor-ß and progesterone receptors under each condition. Oxygen deprivation increased the expression of inducible nitric oxide synthetase (iNOS) and up-regulated selected cytokines and chemokines. Both hormones selectively prevented the induction of pro-inflammatory iNOS, interleukin IL-1ß, and chemokine ligand CCL5, whereas anti-inflammatory IL-10 and protective TREM 2 were up-regulated by sex steroids. Sex hormones abrogated hypoxia-dependent reduction of BV-2 phagocytic activity. We demonstrate that BV-2 microglia cells respond to hypoxia by enhanced pro-inflammatory cytokine secretion and reduced phagocytic activity. This effect is prevented by sex steroids resulting in a switch of BV-2 cells from a pro-inflammatory to a more anti-inflammatory phenotype. Anti-inflammatory effects of gonadal steroids might directly be mediated through hormone-microglia interactions in addition to known effects via astroglial regulation.


Subject(s)
Cell Hypoxia/drug effects , Gonadal Steroid Hormones/pharmacology , Microglia/metabolism , Animals , Chemokine CCL5/metabolism , Chemokines/metabolism , Cytokines/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Mice , Microglia/drug effects , Nitric Oxide Synthase Type II/metabolism
20.
Wound Repair Regen ; 21(1): 44-54, 2013.
Article in English | MEDLINE | ID: mdl-23126541

ABSTRACT

In adults, repair of deeply injured skin wounds results in the formation of scar tissue, whereas in embryos wounds heal almost scar-free. Macrophages are important mediators of wound healing and secrete cytokines and tissue remodeling enzymes. In contrast to host defense mediated by inflammatory M1 macrophages, wound healing and tissue repair involve regulatory M2/M2-like macrophages. Embryonic/fetal macrophages are M2-like, and this may promote scar-free wound healing. In the present study, we asked whether atopical application of ex vivo generated, embryonic stem cell-derived macrophages (ESDM) improve wound healing in mice. ESDM were tested side by side with bone marrow-derived macrophages (BMDM). Compared to BMDM, ESDM resembled a less inflammatory and more M2-like macrophage subtype as indicated by their reduced responsiveness to lipopolysaccharide, reduced expression of Toll-like receptors, and reduced bacterial phagocytosis. Despite this anti-inflammatory phenotype in cell culture, ESDM prolonged the healing of deep skin wounds even more than BMDM. Healed wounds had more scar formation compared to wounds receiving BMDM or cell-free treatment. Our data indicate that atopical application of ex vivo generated macrophages is not a suitable cell therapy of dermal wounds.


Subject(s)
Cicatrix/therapy , Embryonic Stem Cells/transplantation , Inflammation/therapy , Macrophages/metabolism , Skin/pathology , Wound Healing , Wounds and Injuries/therapy , Animals , Cells, Cultured , Cicatrix/pathology , Disease Models, Animal , Flow Cytometry , Humans , Inflammation/pathology , Mice , Mice, Inbred C57BL , Regeneration , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...