Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Ann Neurol ; 87(3): 405-418, 2020 03.
Article in English | MEDLINE | ID: mdl-31900946

ABSTRACT

OBJECTIVE: Leucine-rich glioma-inactivated 1 (LGI1) encephalitis is the second most common antibody-mediated encephalopathy, but insight into the intrathecal B-cell autoimmune response, including clonal relationships, isotype distribution, frequency, and pathogenic effects of single LGI1 antibodies, has remained limited. METHODS: We cloned, expressed, and tested antibodies from 90 antibody-secreting cells (ASCs) and B cells from the cerebrospinal fluid (CSF) of several patients with LGI1 encephalitis. RESULTS: Eighty-four percent of the ASCs and 21% of the memory B cells encoded LGI1-reactive antibodies, whereas reactivities to other brain epitopes were rare. All LGI1 antibodies were of IgG1, IgG2, or IgG4 isotype and had undergone affinity maturation. Seven of the overall 26 LGI1 antibodies efficiently blocked the interaction of LGI1 with its receptor ADAM22 in vitro, and their mean LGI1 signal on mouse brain sections was weak compared to the remaining, non-ADAM22-competing antibodies. Nevertheless, both types of LGI1 antibodies increased the intrinsic cellular excitability and glutamatergic synaptic transmission of hippocampal CA3 neurons in slice cultures. INTERPRETATION: Our data show that the patients' intrathecal B-cell autoimmune response is dominated by LGI1 antibodies and that LGI1 antibodies alone are sufficient to promote neuronal excitability, a basis of seizure generation. Fundamental differences in target specificity and antibody hypermutations compared to the CSF autoantibody repertoire in N-methyl-D-aspartate receptor encephalitis underline the clinical concept that autoimmune encephalitides are very distinct entities. Ann Neurol 2020;87:405-418.


Subject(s)
Antibodies, Monoclonal/pharmacology , Autoantibodies/pharmacology , Intracellular Signaling Peptides and Proteins/immunology , Neurons/physiology , ADAM Proteins/drug effects , Aged , Animals , Antibodies, Monoclonal/cerebrospinal fluid , Autoantibodies/cerebrospinal fluid , CA3 Region, Hippocampal/physiology , Cells, Cultured , Encephalitis/cerebrospinal fluid , Encephalitis/immunology , Female , Hashimoto Disease/cerebrospinal fluid , Hashimoto Disease/immunology , Humans , Immunoglobulin Isotypes , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Middle Aged , Nerve Tissue Proteins/drug effects , Rats , Synaptic Transmission/drug effects
2.
Osteoarthritis Cartilage ; 24(1): 178-187, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26256766

ABSTRACT

OBJECTIVE: To determine whether mandibular condylar cartilage degradation induced by experimentally abnormal occlusion could be ameliorated via systemic administration of strontium or NBD peptide. METHODS: Six-week-old female C57BL/6J mice were used. From the seventh day after mock operation or unilateral anterior crossbite (UAC) treatment, the control and UAC mice were further respectively pharmacologically treated for 2 weeks or 4 weeks of saline (CON + Saline and UAC + Saline groups), SrCl2 (CON + SrCl2 and UAC + SrCl2 groups) or NBD peptide (CON + NBD peptide and UAC + NBD peptide groups). Changes in condylar cartilage and subchondral bone were assessed 21 and 35 days after mock operation or UAC procedure by histology and micro-CT. Real-time PCR and/or immunohistochemistry (IHC) were performed to evaluate changes in expression levels of col2a1, aggrecan, ADAMTS-5, tnf-α, il-1ß, nfkbia, nuclear factor-kappaB phospho-p65 in condylar cartilage, and rankl/rank/opg in both condylar cartilage and subchondral bone. RESULTS: Cartilage degradation with decreased col2a1 and aggrecan expression, and increased ADAMTS-5, tnf-α/il1-ß, nfkbia and NF-κB phospho-p65 was observed in UAC + Saline groups. Subchondral bone loss with increased osteoclast numbers and decreased opg/rankl ratio was found in UAC + Saline groups compared to age-match CON + Saline groups. Cartilage degradation and subchondral bone loss were reversed by treatment of SrCl2 or NBD peptide while the same dosage in control mice induced few changes in condylar cartilage and subchondral bone. CONCLUSIONS: The results demonstrate reverse effect of systemic administration of strontium or NBD peptide on UAC-induced condylar cartilage degradation and subchondral bone loss.


Subject(s)
Cartilage, Articular/drug effects , Malocclusion , Mandibular Condyle/drug effects , Osteoclasts/drug effects , Peptides/pharmacology , RNA, Messenger/drug effects , Strontium/pharmacology , ADAM Proteins/drug effects , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS5 Protein , Aggrecans/drug effects , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Collagen Type II/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Dental Occlusion , Female , I-kappa B Proteins/drug effects , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Immunohistochemistry , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mandibular Condyle/metabolism , Mandibular Condyle/pathology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Osteoclasts/metabolism , Osteoprotegerin/drug effects , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Proteoglycans/drug effects , Proteoglycans/metabolism , RANK Ligand/drug effects , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/drug effects , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24899059

ABSTRACT

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Subject(s)
ADAM Proteins/physiology , Cell Proliferation/physiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/physiology , Glycolysis/physiology , Kidney Tubules, Collecting/pathology , Polycystic Kidney Diseases/physiopathology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/drug effects , ADAM17 Protein , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , ErbB Receptors/physiology , Female , Heparin-binding EGF-like Growth Factor/physiology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiopathology , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Transforming Growth Factor alpha/physiology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
4.
Arterioscler Thromb Vasc Biol ; 32(10): 2444-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22904271

ABSTRACT

OBJECTIVE: Endothelium dysfunction is an initiating factor in atherosclerosis. A disintegrin and metalloproteinase 15 (ADAM 15) is a multidomain metalloprotease recently identified as a regulator of endothelial permeability. However, whether and how ADAM15 contributes to atherosclerosis remains unknown. METHODS AND RESULTS: Genetic ablation of ADAM15 in apolipoprotein E-deficient mice led to a significant reduction in aortic atherosclerotic lesion size (by 52%), plaque macrophage infiltration (by 69%), and smooth muscle cell deposition (by 82%). In vitro studies implicated endothelial-derived ADAM15 in barrier dysfunction and monocyte transmigration across mouse aortic and human umbilical vein endothelial cell monolayers. This role of ADAM15 depended on intact functioning of the cytoplasmic domain, as evidenced in experiments with site-directed mutagenesis targeting the metalloprotease active site (E349A), the disintegrin domain (Arginine-Glycine-Aspartic acid→Threonine-Aspartic acid-Aspartic acid), or the cytoplasmic tail. Further investigations revealed that ADAM15-induced barrier dysfunction was concomitant with dissociation of endothelial adherens junctions (vascular endothelial [VE]-cadherin/γ-catenin), an effect that was sensitive to Src family kinase inhibition. Through small interfering RNA-mediated knockdown of distinct Src family kinase members, c-Src and c-Yes were identified as important mediators of these junctional effects of ADAM15. CONCLUSIONS: These results suggest that endothelial cell-derived ADAM15, signaling through c-Src and c-Yes, contributes to atherosclerotic lesion development by disrupting adherens junction integrity and promoting monocyte transmigration.


Subject(s)
ADAM Proteins/physiology , Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Membrane Proteins/physiology , Signal Transduction/physiology , src-Family Kinases/physiology , ADAM Proteins/drug effects , ADAM Proteins/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , CSK Tyrosine-Protein Kinase , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/pathology , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Monocytes/physiology , Proto-Oncogene Proteins c-yes/drug effects , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/physiology , RNA, Small Interfering/pharmacology , src-Family Kinases/drug effects , src-Family Kinases/genetics
5.
BMC Gastroenterol ; 13: 155, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24172289

ABSTRACT

BACKGROUND: Ursodeoxycholic acid (UDCA) is used to treat primary biliary cirrhosis, intrahepatic cholestasis, and other cholestatic conditions. Although much has been learned about the molecular basis of the disease pathophysiology, our understanding of the effects of UDCA remains unclear. Possibly underlying its cytoprotective, anti-apoptotic, anti-oxidative effects, UDCA was reported to regulate the expression of TNFα and other inflammatory cytokines. However, it is not known if this effect involves also modulation of ADAM family of metalloproteinases, which are responsible for release of ectodomains of inflammatory cytokines from the cell surface. We hypothesized that UDCA modulates ADAM17 activity, resulting in amelioration of cholestasis in a murine model of bile duct ligation (BDL). METHODS: The effect of UDCA on ADAM17 activity was studied using the human liver hepatocellular carcinoma cell line HepG2. Untransfected cells or cells ectopically expressing human ADAM17 were cultured with or without UDCA and further activated using phorbol-12-myristate-13-acetate (PMA). The expression and release of ADAM17 substrates, TNFα, TGFα, and c-Met receptor (or its soluble form, sMet) were evaluated using ELISA and quantitative real-time (qRT) PCR. Immunoblotting analyses were conducted to evaluate expression and activation of ADAM17 as well as the level of ERK1/2 phosphorylation after UDCA treatment. The regulation of tissue inhibitor of metalloproteinases-1 (TIMP-1) by UDCA was studied using zymography and qRT-PCR. A mouse model of acute cholestasis was induced by common BDL technique, during which mice received daily orogastric gavage with either UDCA or vehicle only. Liver injury was quantified using alkaline phosphatase (ALP), relative liver weight, and confirmed by histological analysis. ADAM17 substrates in sera were assessed using a bead multiplex assay. RESULTS: UDCA decreases amount of shed TNFα, TGFα, and sMet in cell culture media and the phosphorylation of ERK1/2. These effects are mediated by the reduction of ADAM17 activity in PMA stimulated cells although the expression ADAM17 is not affected. UDCA reduced the level of the mature form of ADAM17. Moreover, UDCA regulates the expression of TIMP-1 and gelatinases activity in PMA stimulated cells. A BDL-induced acute cholangitis model was characterized by increased relative liver weight, serum levels of ALP, sMet, and loss of intracellular glycogen. UDCA administration significantly decreased ALP and sMet levels, and reduced relative liver weight. Furthermore, hepatocytes of UDCA-treated animals retained their metabolic activity as evidenced by the amount of glycogen storage. CONCLUSIONS: The beneficial effect of UDCA appears to be mediated in part by the inhibition of ADAM17 activation and, thus, the release of TNFα, a strong pro-inflammatory factor. The release of other ADAM17 substrates, TGFα and sMet, are also regulated this way, pointing to a general impact on the release of ADAM17 substrates, which are pivotal for liver regeneration and function. In parallel, UDCA upregulates TIMP-1 that in turn inhibits matrix metalloproteinases, which destroy the hepatic ECM in diseased liver. This control of extracellular matrix turnover represents an additional beneficial path of UDCA treatment.


Subject(s)
ADAM Proteins/drug effects , Cholagogues and Choleretics/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Ursodeoxycholic Acid/pharmacology , ADAM17 Protein , Animals , Bile Ducts/surgery , Cholestasis , Hep G2 Cells , Humans , Ligation , MAP Kinase Signaling System/drug effects , Mice , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/metabolism , Transforming Growth Factor alpha/drug effects , Transforming Growth Factor alpha/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Toxicol Appl Pharmacol ; 263(1): 61-72, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22687607

ABSTRACT

Mer receptor tyrosine kinase (Mer) regulates macrophage activation and promotes apoptotic cell clearance. Mer activation is regulated through proteolytic cleavage of the extracellular domain. To determine if membrane-bound Mer is cleaved during bleomycin-induced lung injury, and, if so, how preventing the cleavage of Mer enhances apoptotic cell uptake and down-regulates pulmonary immune responses. During bleomycin-induced acute lung injury in mice, membrane-bound Mer expression decreased, but production of soluble Mer and activity as well as expression of disintegrin and metalloproteinase 17 (ADAM17) were enhanced . Treatment with the ADAM inhibitor TAPI-0 restored Mer expression and diminished soluble Mer production. Furthermore, TAPI-0 increased Mer activation in alveolar macrophages and lung tissue resulting in enhanced apoptotic cell clearance in vivo and ex vivo by alveolar macrophages. Suppression of bleomycin-induced pro-inflammatory mediators, but enhancement of hepatocyte growth factor induction were seen after TAPI-0 treatment. Additional bleomycin-induced inflammatory responses reduced by TAPI-0 treatment included inflammatory cell recruitment into the lungs, levels of total protein and lactate dehydrogenase activity in bronchoalveolar lavage fluid, as well as caspase-3 and caspase-9 activity and alveolar epithelial cell apoptosis in lung tissue. Importantly, the effects of TAPI-0 on bleomycin-induced inflammation and apoptosis were reversed by coadministration of specific Mer-neutralizing antibodies. These findings suggest that restored membrane-bound Mer expression by TAPI-0 treatment may help resolve lung inflammation and apoptosis after bleomycin treatment.


Subject(s)
Acute Lung Injury/prevention & control , Phagocytosis/drug effects , Receptor Protein-Tyrosine Kinases/physiology , ADAM Proteins/drug effects , ADAM Proteins/metabolism , ADAM17 Protein , Acute Lung Injury/chemically induced , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Bleomycin/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase 9/drug effects , DNA Damage/drug effects , Dipeptides/pharmacology , Enzyme-Linked Immunosorbent Assay , Hydroxamic Acids/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Eur Cell Mater ; 23: 41-57, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22287113

ABSTRACT

This study investigated whether treatment of articular cartilage chondrocytes with a combination of oncostatin M (OSM) and interleukin-1 (IL-1) could induce a degradative phenotype that was mediated through the protein kinase R (PKR) signalling pathway. High-density monolayer cultures of full depth, bovine chondrocytes were treated with a combination of OSM and IL-1 (OSM+IL-1) for 7 days. To inhibit the activation of PKR, a pharmacological inhibitor of PKR was added to duplicate cultures. Pro- and active matrix metalloproteinase-9 (MMP9) and MMP9 mRNA were significantly upregulated by OSM+IL-1 through a PKR dependent mechanism. ADAMTS4 and ADAMTS5 mRNA were also upregulated by OSM+IL-1. The upregulation of ADAMTS4 and ADAMTS5 were, in part, mediated through PKR. OSM+IL-1 resulted in a loss of type II collagen, which could not be rescued by PKR inhibition. OSM+IL-1 reduced the expression of COL2A1 (type II collagen), COL9A1 (type IX collagen), COL11A1 (type XI collagen), and ACAN (aggrecan) mRNAs. Expression of type II and XI collagen and aggrecan was reduced further when PKR was inhibited. OSM+IL-1 resulted in an 11-fold increase in TNFa mRNA which was, in part, mediated through the PKR pathway. This study demonstrates, for the first time, that a number of catabolic and pro-inflammatory effects known to be important in human arthritis and induced by OSM and IL-1, are mediated by the PKR signalling pathway.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Interleukin-1/administration & dosage , Oncostatin M/administration & dosage , Oncostatin M/metabolism , eIF-2 Kinase/metabolism , ADAM Proteins/drug effects , ADAM Proteins/metabolism , Animals , Cattle , Chondrocytes/drug effects , Collagen Type II/drug effects , Collagen Type II/metabolism , Drug Combinations , Enzyme Inhibitors , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Primary Cell Culture , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/antagonists & inhibitors
8.
Nephrol Dial Transplant ; 27(5): 2114-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22015440

ABSTRACT

BACKGROUND: Interstitial fibrosis and tubular atrophy (IF/TA) is an important cause of renal function loss and ischaemia-reperfusion (I/R) injury is considered to play an important role in its pathophysiology. The aim of the present study was to investigate the role of a disintegrin and metalloproteinase 17 (ADAM17) in human renal allograft disease and in experimental I/R injury of the kidney. METHODS: We studied the expression of ADAM17 messenger RNA (mRNA) in IF/TA and control kidneys by reverse transcription-polymerase chain reaction and in situ hybridization. Moreover, we assessed ADAM17-mediated heparin-binding epidermal growth factor (HB-EGF) shedding in immortalized human cells. Finally, we studied the effect of pharmacological ADAM17 inhibition in a model of renal I/R injury in rats. RESULTS: ADAM17 mRNA was up-regulated in IF/TA when compared to control kidneys. In normal kidneys, ADAM17 mRNA was weakly expressed in proximal tubules, peritubular capillaries, glomerular endothelium and parietal epithelium. In IF/TA, tubular, capillary and glomerular ADAM17 expression was strongly enhanced with de novo expression in the mesangium. In interstitial fibrotic lesions, we observed co-localization of ADAM17 with HB-EGF protein. In vitro, inhibition of ADAM17 with TNF484 resulted in a dose-dependent reduction of HB-EGF shedding in phorbol 12-myrisate 13-acetate-stimulated cells and non-stimulated cells. In vivo, ADAM17 inhibition significantly reduced the number of glomerular and interstitial macrophages at Day 4 of reperfusion. CONCLUSIONS: In conclusion, HB-EGF co-expresses with ADAM17 in renal interstitial fibrosis, suggesting a potential interaction in IF/TA. Targeting ADAM17 to reduce epidermal growth factor receptor phosphorylation could be a promising way of intervention in human renal disease.


Subject(s)
ADAM Proteins/metabolism , Kidney Transplantation , Kidney/metabolism , Kidney/pathology , Reperfusion Injury/metabolism , Up-Regulation , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/drug effects , ADAM17 Protein , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Atrophy , Cells, Cultured , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Fibrosis , Heparin-binding EGF-like Growth Factor , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/drug effects , Male , Middle Aged , Models, Animal , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reperfusion Injury/pathology , Young Adult
9.
Arch Toxicol ; 86(10): 1537-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22581348

ABSTRACT

Human exposure to PM(2.5) (particulate matter with an aerodynamic diameter below 2.5 µm) is known to be responsible for airway inflammation and may also induce airway remodelling. In respiratory epithelial cells exposed to PM(2.5), releases of pro-inflammatory cytokines such as granulocyte macrophage-colony stimulating factor (GM-CSF) and growth factor ligands of the epidermal growth factor receptor (EGFR) are increased. The present study aimed at determining the involvement of EGFR ligands by autocrine effects in PM(2.5)-induced GM-CSF release. PM(2.5) exposure triggers GM-CSF release by human bronchial epithelial (HBE) cells. This release is dependent on EGFR activation by ligand binding as it is inhibited by AG1478, an inhibitor of EGFR tyrosine kinase activity as well as by a neutralizing anti-EGFR antibody. The use of conditioned medium from cells previously exposed to PM(2.5) demonstrates that PM(2.5)-exposed cells release soluble EGFR ligands able to induce GM-CSF release by an autocrine manner. It was further demonstrated by inhibiting tumour-necrosis factor-alpha converting enzyme (TACE) that is involved in some EGFR ligand shedding. TAPI-2 and GM-6001, two TACE inhibitors, prevented the PM(2.5)-induced GM-CSF release as well as the silencing of TACE by siRNA. We provide evidence that the pro-inflammatory response induced by PM(2.5) exposure on HBE cells, results from an autocrine effect of EGFR ligands released by TACE activity. This autocrine loop by eliciting and sustaining inflammation could contribute to exacerbation of airway remodelling in respiratory-compromised individuals.


Subject(s)
Epithelial Cells/drug effects , ErbB Receptors/drug effects , Inflammation/chemically induced , Particulate Matter/toxicity , ADAM Proteins/drug effects , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Airway Remodeling/drug effects , Autocrine Communication/drug effects , Bronchi/cytology , Bronchi/drug effects , Cell Line , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Gene Silencing , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/pathology , Particle Size , Quinazolines/pharmacology , RNA, Small Interfering/metabolism , Tyrphostins/pharmacology
10.
Cell Tissue Res ; 340(1): 179-88, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20217136

ABSTRACT

Pro-inflammatory cytokines induce meniscal matrix degradation and inhibition of endogenous repair mechanisms, but the pathogenic mechanisms behind this are mostly unknown. Therefore, we investigated details of interleukin-1 (IL-1alpha)-induced aggrecan turnover in mature meniscal tissue explants. Fibro-cartilagenous disks (3 mm diameter x 1 mm thickness) were isolated from the central, weight-bearing region of menisci from 2-year-old cattle. After 3 or 6 days of IL-1alpha-treatment, GAG loss (DMMB assay), biosynthetic activity ([(35)SO(4)]-sulfate and [(3)H]-proline incorporation), gene expression (quantitative RT-PCR) and the abundance (zymography, Western blot) of matrix-degrading enzymes and specific aggrecan products were determined. Meniscal fibrocartilage had a 4-fold lower GAG content (per wet weight) than adjacent articular cartilage, and expressed MMPs-1, -2, -3 and ADAMTS4 constitutively, whereas ADAMTS5 m-RNA was essentially undetectable. Significant IL-1 effects were a decrease in biosynthetic activity, an increase in GAG release and in the expression/abundance of MMP-2, MMP-3 and ADAMTS4. Fresh tissue contained aggrecan core protein products similar to those previously described for bovine articular cartilage of this age. IL-1 induced the release of aggrecanase-generated CS-substituted products including both high (>250 kDa) and low molecular weight (about 75 kDa) species. TIMP-3 (but not TIMP-1 and -2 or a broad spectrum MMP inhibitor) inhibited IL-1-dependent GAG loss. In addition, IL-1 induced the release of preformed pools of three known G1-bearing products. We conclude that aggrecanases are responsible for IL-1-stimulated GAG release from meniscal explants, and that IL-1 also stimulates release of G1-bearing products, by a process possibly involving hyaluronan fragmentation.


Subject(s)
Aggrecans/metabolism , Arthritis/immunology , Glycosaminoglycans/metabolism , Inflammation Mediators/metabolism , Interleukin-1alpha/metabolism , Menisci, Tibial/immunology , ADAM Proteins/drug effects , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS4 Protein , Aggrecans/drug effects , Animals , Arthritis/metabolism , Arthritis/physiopathology , Calpain/drug effects , Calpain/genetics , Calpain/metabolism , Cattle , Endopeptidases/drug effects , Endopeptidases/genetics , Endopeptidases/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Hyaluronic Acid/metabolism , Inflammation Mediators/pharmacology , Interleukin-1alpha/pharmacology , Matrix Metalloproteinases/drug effects , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Menisci, Tibial/drug effects , Menisci, Tibial/metabolism , Models, Biological , Procollagen N-Endopeptidase/drug effects , Procollagen N-Endopeptidase/genetics , Procollagen N-Endopeptidase/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-3/drug effects , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
11.
Osteoarthritis Cartilage ; 18(3): 455-63, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19909832

ABSTRACT

OBJECTIVE: Glucosamine has been previously shown to suppress cartilage aggrecan catabolism in explant cultures. We determined the effect of glucosamine on ADAMTS5 (a disintegrin-like and metalloprotease domain (reprolysin type) with thrombospondin type-1 motifs 5), a major aggrecanase in osteoarthritis, and investigated a potential mechanism underlying the observed effects. DESIGN: HEK293F and CHO-K1 cells transiently transfected with ADAMTS5 cDNA were treated with glucosamine or the related hexosamine mannosamine. Glucosamine effects on FURIN transcription were determined by quantitative RT-PCR. Effects on furin-mediated processing of ADAMTS5 zymogen, and aggrecan processing by glucosamine-treated cells, were determined by western blotting. Post-translational modification of furin and N-glycan deficient furin mutants generated by site-directed mutagenesis was analyzed by western blotting, and the mutants were evaluated for their ADAMTS5 processing ability in furin-deficient CHO-RPE.40 cells. RESULTS: Ten mM glucosamine and 5-10mM mannosamine reduced excision of the ADAMTS5 propeptide, indicating interference with the propeptide excision mechanism, although mannosamine compromised cell viability at these doses. Although glucosamine had no effect on furin mRNA levels, western blot of furin from glucosamine-treated cells suggested altered post-translational modification. Glucosamine treatment led to decreased glycosylation of cellular furin, with reduced furin autoactivation as the consequence. Recombinant furin treated with peptide N-glycanase F had reduced activity against a synthetic peptide substrate. Indeed, site-directed mutagenesis of two furin N-glycosylation sites, Asn(387) and Asn(440), abrogated furin activation and this mutant was unable to rescue ADAMTS5 processing in furin-deficient cells. CONCLUSIONS: Ten mM glucosamine reduces excision of the ADAMTS5 propeptide via interference with post-translational modification of furin and leads to reduced aggrecanase activity of ADAMTS5.


Subject(s)
ADAM Proteins/drug effects , Furin/drug effects , Glucosamine/metabolism , ADAMTS5 Protein , Blotting, Western , Cells, Cultured , Humans , Protein Processing, Post-Translational , Reverse Transcriptase Polymerase Chain Reaction , Statistics as Topic
12.
Osteoarthritis Cartilage ; 18(1): 106-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786147

ABSTRACT

OBJECTIVE: Although most studies have focused on the cholesterol-lowering activity of stigmasterol, other bioactivities have been ascribed to this plant sterol compound, one of which is a potential anti-inflammatory effect. To investigate the effects of stigmasterol, a plant sterol, on the inflammatory mediators and metalloproteinases produced by chondrocytes. METHOD: We used a model of newborn mouse chondrocytes and human osteoarthritis (OA) chondrocytes in primary culture stimulated with or without IL-1beta (10 ng/ml), for 18 h. Cells were pre-incubated for 48 h with stigmasterol (20 microg/ml) compared to untreated cells. We initially investigated the presence of stigmasterol in chondrocyte, compared to other phytosterols. We then assessed the role of stigmasterol on the expression of various genes involved in inflammation (IL-6) and cartilage turn-over (MMP-3, -13, ADAMTS-4, -5, type II collagen, aggrecan) by quantitative Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR). Additional experiments were carried out to monitor the production of MMP-3 and prostaglandin E2 (PGE(2)) by specific immuno-enzymatic assays. We eventually looked at the role of stigmasterol on NF-kappaB activation by western blot, using an anti-IkappaBalpha antibody. RESULTS: After 18 h of IL-1beta treatment, MMP-3, MMP-13, ADAMTS-4, but not ADAMTS-5 RNA expression were elevated, as well as MMP-3 and PGE(2) protein levels in mouse and human chondrocytes. Type II collagen and aggrecan mRNA levels were significatively reduced. Pre-incubation of stigmasterol to IL-1beta-treated cells significantly decreased these effects described above (significant reduction of MMP-3 mRNA in human and mouse, MMP-3 protein in mouse, MMP-13 mRNA in mouse and human, ADAMTS-4 mRNA in human, PGE(2) protein in human and mouse) Finally, stigmasterol was capable of counteracting the IL-1beta-induced NF-kappaB pathway. CONCLUSION: This study shows that stigmasterol inhibits several pro-inflammatory and matrix degradation mediators typically involved in OA-induced cartilage degradation, at least in part through the inhibition of the NF-kappaB pathway. These promising results justify further ex vivo and in vivo investigations with stigmasterol.


Subject(s)
ADAM Proteins/metabolism , Chondrocytes/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Osteoarthritis, Knee/metabolism , Stigmasterol/pharmacology , ADAM Proteins/drug effects , ADAMTS5 Protein , Aged , Aged, 80 and over , Animals , Blotting, Western , Cell Death , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chondrocytes/immunology , Chondrocytes/metabolism , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-1beta/pharmacology , L-Lactate Dehydrogenase/analysis , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 3/drug effects , Mice , Middle Aged , Models, Animal , NF-kappa B/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Crit Care Med ; 36(10): 2878-87, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18828196

ABSTRACT

BACKGROUND: Thrombocytopenia-associated multiple organ failure (TAMOF) is a poorly understood syndrome in critically ill children. A disintegrin and metalloprotease with thrombospondin motifs (ADAMTS-13), formerly known as von Willebrand factor (VWF) cleaving protease, is decreased in adults with VWF-mediated thrombotic microangiopathy, and intensive plasma exchange (PEx) both replenishes ADAMTS-13 and improves outcome in these patients. OBJECTIVES: To determine whether: 1) critically ill children with TAMOF syndrome have decreased ADAMTS-13 activity, 2) ADAMTS-13 activity correlates with platelet counts and VWF antigen, 3) the autopsies from patients who died with reduced ADAMTS-13 activity have VWF-rich microthrombi, and 4) intensive PEx will restore ADAMTS-13 activity and facilitate organ failure resolution. DESIGN: First study: observational. Second study: randomized control trial. SETTING: Single center university pediatric intensive care unit. PATIENTS: First study: thirty-seven consecutive children (17 males and 20 females; ages ranging from 9 days to 23 years) identified with > or = 2 organs dysfunction were enrolled. Seventy-six percent of these children had thrombocytopenia (platelet counts < 100,000/mm3). Five additional critically ill children without MOF were also enrolled. In the second study, children with severe TAMOF (platelet counts < 100,000/mm3 and > 3 organ failure) were randomized to PEx or standard therapy. Primary physicians and parents agreed to enrollment in 10 of the 20 eligible patients with ages ranging from 1 year to 18 years. Five patients received PEx and 5 patients received standard therapy. RESULTS: First study: children with TAMOF (n = 28) had decreased ADAMTS-13 activity, but similar plasminogen activator inhibitor-1 activity and prothrombin time compared to children with MOF without thrombocytopenia (n = 9, p < 0.05). All non-survivors (n = 7) had TAMOF, reduced ADAMTS-13 activity, and VWF-rich microvascular thromboses at autopsy. In the second study, PEx (n = 5, median 12 days, 4-28 days) restored ADAMTS-13 activity and organ function, compared to standard therapy (n = 5, p < 0.05). CONCLUSIONS: Children with TAMOF syndrome can have VWF-mediated thrombotic microangiopathy. Similar to adult experience, PEx can replenish ADAMTS-13 activity and reverse organ failure.


Subject(s)
ADAM Proteins/blood , Multiple Organ Failure/therapy , Plasma Exchange/methods , Thrombocytopenia/therapy , ADAM Proteins/drug effects , ADAMTS13 Protein , Adolescent , Adult , Age Factors , Analysis of Variance , Biomarkers/blood , Child , Child, Preschool , Critical Illness/mortality , Critical Illness/therapy , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Logistic Models , Male , Multiple Organ Failure/blood , Multiple Organ Failure/complications , Multiple Organ Failure/mortality , Reference Values , Risk Assessment , Statistics, Nonparametric , Survival Rate , Thrombocytopenia/blood , Thrombocytopenia/complications , Thrombocytopenia/mortality , Treatment Outcome
14.
Int J Biochem Cell Biol ; 105: 123-133, 2018 12.
Article in English | MEDLINE | ID: mdl-30343150

ABSTRACT

The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.


Subject(s)
Ephrins/drug effects , Ephrins/metabolism , Receptors, Eph Family/drug effects , Receptors, Eph Family/metabolism , ADAM Proteins/drug effects , ADAM Proteins/metabolism , Antibodies/chemistry , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Binding Sites , Drug Development , Ephrins/chemistry , Humans , Models, Biological , Models, Molecular , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Eph Family/genetics , Signal Transduction/drug effects
15.
Neuroscience ; 150(2): 386-95, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17945434

ABSTRACT

Alpha-secretase (alpha-secretase), cleaves the amyloid precursor protein (APP) within the amyloid-beta (Abeta) sequence, resulting in the release of a secreted fragment of APP (alphaAPPs) and precluding Abeta generation. We investigated the effects of the acetylcholinesterase inhibitor, huperzine A (Hup A), on APP processing and Abeta generation in human neuroblastoma SK-N-SH cells overexpressing wild-type human APP695. Hup A dose-dependently (0-10 microM) increased alphaAPPs release. Therefore, we evaluated two alpha-secretase candidates, a disintegrin and metalloprotease (ADAM) 10 and ADAM17 in Hup A-induced non-amyloidogenic APP metabolism. Hup A enhanced the level of ADAM10, and the inhibitor of tumor necrosis factor-alpha converting enzyme (TACE)/ADAM17 inhibited the Hup A-induced rise in alphaAPPs levels, further suggesting Hup A directed APP metabolism toward the non-amyloidogenic alpha-secretase pathway. Hup A had no effect on Abeta generation in this cell line. The steady-state levels of full-length APP and cell viability were unaffected by Hup A. Alpha-APPs release induced by Hup A treatment was significantly reduced by muscarinic acetylcholine receptor antagonists (particularly by an M1 antagonist), protein kinase C (PKC) inhibitors, GF109203X and calphostin C, and the mitogen-activated kinase kinase (MEK) inhibitors, U0126 and PD98059. Furthermore, Hup A markedly increased the phosphorylation of p44/p42 mitogen-activated protein (MAP) kinase, which was blocked by treatment with U0126 and PD98059. In addition, Hup A inhibited acetylcholinesterase activity by 20% in neuroblastoma cells. Our results indicate that the activation of muscarinic acetylcholine receptors, PKC and MAP kinase may be involved in Hup A-induced alphaAPPs secretion in neuroblastoma cells and suggest multiple pharmacological mechanisms of Hup A regarding the treatment of Alzheimer's disease (AD).


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/drug effects , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Sesquiterpenes/pharmacology , ADAM Proteins/drug effects , ADAM Proteins/metabolism , ADAM10 Protein , ADAM17 Protein , Alkaloids , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/physiology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Muscarinic Antagonists/pharmacology , Neuroblastoma , Neurons/metabolism , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism
16.
ACS Chem Neurosci ; 7(1): 40-5, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26559179

ABSTRACT

The ADAM family of metalloproteases cleave a diverse range of transmembrane substrates, resulting in the release of their soluble ectodomains. This process of protein shedding, termed α-secretase processing, is involved in many facets of both normal and disease related cellular function. While the processing of substrates has been well documented, the regulation and trafficking of the ADAMs are less well understood. Tools that allow for the study of ADAMs under their native environment will allow for a better understanding of their regulation and activity. Here we describe the design and evaluation of a novel fluorescent analogue of a well-characterized ADAM inhibitor, Batimastat. This probe exhibited similar activity for inhibiting α-secretase processing in cells as did Batimastat. Importantly, this probe specifically labeled ADAMs fluorescently in both fixed and living cells, enabling the possibility to study the trafficking of α-secretase proteins in a dynamic environment.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Phenylalanine/analogs & derivatives , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , ADAM Proteins/drug effects , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protease Inhibitors/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thiophenes/chemistry , Transfection
17.
Int Immunopharmacol ; 29(2): 863-868, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371861

ABSTRACT

Hypothemycin, a resorcylic acid lactone polyketide, has been shown to inhibit oncogenic ras-transformation and T cell activation. In the present study, we investigated the effect of hypothemycin on tumor necrosis factor-α (TNF-α) production in macrophages and the molecular mechanisms involved in this effect. Hypothemycin potently suppressed the TNF-α production without affecting nitric oxide production in lipopolysaccharide (LPS)-stimulated macrophages. However, hypothemycin had no effect on the activity of TNF-α-converting enzyme, a key enzyme for converting membrane-bound pro-TNF-α into soluble TNF-α. Further study demonstrated that the stability of TNF-α mRNA was decreased by hypothemycin treatment. In addition, hypothemycin suppressed LPS-induced phosphorylation of p38 MAPK and ERK. Moreover, knockdown of tristetraprolin (TTP), which is an important trans-acting regulator of TNF-α mRNA stability and downstream target of p38 MAPK and ERK, reversed hypothemycin-mediated inhibition of TNF-α mRNA expression. Collectively, our results suggest that hypothemycin suppresses TNF-α production by TTP-dependent destabilization of TNF-α mRNA and this is mediated, at least in part, by blocking the activation of p38 MAPK and ERK.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/metabolism , RNA Stability/drug effects , Tristetraprolin/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Zearalenone/analogs & derivatives , ADAM Proteins/drug effects , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Down-Regulation/drug effects , Humans , Macrophages/drug effects , Mice , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Zearalenone/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
18.
Invest Ophthalmol Vis Sci ; 56(9): 5162-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26244291

ABSTRACT

PURPOSE: The a-disintegrin-and-metalloprotease (ADAM) family proteins are widely expressed in the different layers of the retina throughout development. The effect of ADAM proteins on the epithelial-to-mesenchymal transition (EMT) in proliferative vitreoretinopathy (PVR) or AMD is yet to be elucidated. In this study we used Epstein-Barr virus (EBV)-transformed adult retinal pigment epithelial (ARPE) cells to investigate how sorafenib, a multikinase inhibitor, modulates ADAM proteins to control EMT. METHODS: Epithelial to mesenchymal transition and related mechanisms in EBV-infected ARPE cells were determined by RT-PCR, Western blot, invasion assay, ELISA assay, and gene silencing with siRNA. RESULTS: Mesenchymal-like ARPE/EBV cells exhibited considerably increased cellular migration and invasion compared with ARPE cells and produced EMT-related cytokines. Sorafenib significantly inhibited production of TGF-ß1, VEGF, IL-6, IL-8, MCP-1, and TNF-α and blocked the activation of migration-related signaling molecules, such as HIF-1α, p-STAT3, MMP2, and Ang-1. The expression of mature ADAM10, ADAM17, and cleaved Notch 1 proteins in ARPE/EBV cells was downregulated after treatment with sorafenib through the regulatory activity of nardilysin (NRD-1). Gene silencing of NRD-1 in ARPE/EBV cells attenuated secretion of EMT-related cytokines and expression of ADAM10 and 17 and upregulated epithelial markers. CONCLUSIONS: Sorafenib controls the mesenchymal characteristics of EBV-infected ARPE cells. Nardilysin and ADAM family proteins might be new targets for the prevention or control of EMT in retinal diseases.


Subject(s)
ADAM Proteins/genetics , Amyloid Precursor Protein Secretases/genetics , DNA/genetics , Gene Expression Regulation/drug effects , Herpesvirus 4, Human , Membrane Proteins/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Retinal Pigment Epithelium/metabolism , ADAM Proteins/biosynthesis , ADAM Proteins/drug effects , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/drug effects , Cell Movement , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Immunoblotting , Membrane Proteins/biosynthesis , Membrane Proteins/drug effects , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sorafenib , Tumor Necrosis Factor-alpha , Vitreoretinopathy, Proliferative/genetics , Vitreoretinopathy, Proliferative/pathology , Vitreoretinopathy, Proliferative/virology
19.
Int J Pharm ; 416(2): 486-92, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21382458

ABSTRACT

UNLABELLED: Encapsulation of glucocorticoids into long-circulating liposomes provides targeting of these drugs to the inflamed synovium in experimental arthritis and thereby strongly improves their therapeutic index. The goal of this study was to evaluate the effect and mechanisms of intravenous liposomal delivery of prednisolone phosphate (Lip-PLP) on protease mediated cartilage destruction during murine antigen-induced arthritis (AIA). Mice treated with a single injection of Lip-PLP showed a pronounced suppression of synovial immune cell infiltration compared to control, PBS-treated mice. Liposomal PLP also significantly suppressed interleukin 1ß (3.6 fold) in the synovium, but not in the blood serum. Furthermore, expression of the proteases MMP-3, -9, -13 and -14 and ADAMTS-4 and -5 was suppressed by Lip-PLP in the synovium, but not within the articular cartilage of the femur and tibia, demonstrating the specific action of Lip-PLP on the synovium. Lip-PLP is phagocytosed by macrophages in vitro and suppresses their expression of IL-1ß and MMPs after LPS activation. Moreover, Lip-PLP suppresses destruction of the cartilage matrix during AIA mediated by active MMPs and ADAMTS, as assessed by immunostaining of their respective neoepitopes VDIPEN and NITEGE in various cartilage layers of the knee joint. CONCLUSION: liposomal delivery of glucocorticoids protects against cartilage matrix destruction during experimental arthritis by inhibiting protease expression and activity in the inflamed synovium.


Subject(s)
Arthritis, Experimental/drug therapy , Drug Delivery Systems , Glucocorticoids/pharmacology , Prednisolone/analogs & derivatives , ADAM Proteins/drug effects , ADAM Proteins/metabolism , Animals , Antigens/toxicity , Arthritis, Experimental/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Delayed-Action Preparations , Glucocorticoids/administration & dosage , Interleukin-1beta/metabolism , Liposomes , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Prednisolone/administration & dosage , Prednisolone/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/pathology
20.
Curr Pharm Des ; 15(20): 2319-35, 2009.
Article in English | MEDLINE | ID: mdl-19601834

ABSTRACT

As a metalloproteinase specialized in releasing membrane-tethered proteins, A Disintegrin and A Metalloproteinase 17 (ADAM17), also known as Tumor necrosis factor-alpha Converting Enzyme (TACE) or less commonly CD156q, has received more than its share of attention. This is mainly because major contemporary pathologies like cancer, inflammatory and vascular diseases seem to be connected to its cleavage abilities. The involvement in such a broad spectrum of diseases is due to the large variety of substrates that ADAM17 is able to cut. ADAM17 can activate growth factors or inactivate receptors by shedding their extracellular domain from the cell membrane. Similarly, it can detach cells by cleaving cell adhesion molecules. Some of these proteolytic events are part of cleavage cascades known as Regulated Intramembrane Proteolysis and lead to intracellular signaling. It is therefore clear that ADAM17 literally fulfills a key role in diverse processes and pathologies, making it a prime target for developing therapies. Here we review the role of ADAM17 in health and disease and highlight the problems to overcome for ADAM17 to mature towards a therapeutically valuable target.


Subject(s)
ADAM Proteins/drug effects , ADAM Proteins/chemistry , ADAM17 Protein , Humans , Inflammation/drug therapy , Neoplasms/drug therapy , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL