Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 17(1): 153, 2020 May 09.
Article in English | MEDLINE | ID: mdl-32386505

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease which results from the invasion of the brain by activated immune cells across the endothelial cells (ECs) of the blood-brain barrier (BBB), due to loss of immune self-tolerance. Many reports define the metabolic profile of immune cells in MS, however little is known about the metabolism of the BBB ECs during the disease. We aim to determine whether circulating factors in MS induce metabolic alterations of the BBB ECs compared to a healthy state, which can be linked with disruption of BBB integrity and subsequent immune cell extravasation. METHODS AND RESULTS: In this report, we used an in vitro model to study the effect of sera from naïve-to-treatment, relapsing-remitting MS (RRMS) patients on the human brain microvascular endothelium, comparing effects to age/sex-matched healthy donor (HD) sera. Our data show that RRMS serum components affect brain endothelial cells by impairing intercellular tightness through the down-modulation of occludin and VE-cadherin, and facilitating immune cell extravasation through upregulation of intercellular adhesion molecules (ICAM-1) and P-glycoprotein (P-gp). At a metabolic level, the treatment of the endothelial cells with RRMS sera reduced their glycolytic activity (measured through the extracellular acidification rate-ECAR) and oxygen consumption rate (oxidative phosphorylation rate-OCR). Such changes were associated with the down-modulation of endothelial glucose transporter 1 (GLUT-1) expression and by altered mitochondrial membrane potential. Higher level of reactive oxygen species released from the endothelial cells treated with RRMS sera indicate a pro-inflammatory status of the cells together with the higher expression of ICAM-1, endothelial cell cytoskeleton perturbation (stress fibres) as well as disruption of the cytoskeleton signal transduction MSK1/2 and ß-catenin phosphorylation. CONCLUSIONS: Our data suggest that circulating factors present in RRMS patient serum induce physiological and biochemical alterations to the BBB, namely reducing expression of essential tightness regulators, as well as reduced engagement of glycolysis and alteration of mitochondrial potential. As these last changes have been linked with alterations in nutrient usage and metabolic function in immune cells; we propose that the BBB endothelium of MS patients may similarly undergo metabolic dysregulation, leading to enhanced permeability and increased disease susceptibility.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelium, Vascular/metabolism , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/immunology , Adult , Capillary Permeability/immunology , Cells, Cultured , Female , Humans , Male , Transendothelial and Transepithelial Migration/immunology
2.
Brain Behav Immun ; 51: 212-222, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26321046

ABSTRACT

Sex differences have been widely reported in neuroinflammatory disorders, focusing on the contributory role of estrogen. The microvascular endothelium of the brain is a critical component of the blood-brain barrier (BBB) and it is recognized as a major interface for communication between the periphery and the brain. As such, the cerebral capillary endothelium represents an important target for the peripheral estrogen neuroprotective functions, leading us to hypothesize that estrogen can limit BBB breakdown following the onset of peripheral inflammation. Comparison of male and female murine responses to peripheral LPS challenge revealed a short-term inflammation-induced deficit in BBB integrity in males that was not apparent in young females, but was notable in older, reproductively senescent females. Importantly, ovariectomy and hence estrogen loss recapitulated an aged phenotype in young females, which was reversible upon estradiol replacement. Using a well-established model of human cerebrovascular endothelial cells we investigated the effects of estradiol upon key barrier features, namely paracellular permeability, transendothelial electrical resistance, tight junction integrity and lymphocyte transmigration under basal and inflammatory conditions, modeled by treatment with TNFα and IFNγ. In all cases estradiol prevented inflammation-induced defects in barrier function, action mediated in large part through up-regulation of the central coordinator of tight junction integrity, annexin A1. The key role of this protein was then further confirmed in studies of human or murine annexin A1 genetic ablation models. Together, our data provide novel mechanisms for the protective effects of estrogen, and enhance our understanding of the beneficial role it plays in neurovascular/neuroimmune disease.


Subject(s)
Annexin A1/physiology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Estradiol/physiology , Inflammation/immunology , Inflammation/physiopathology , Lymphocytes/physiology , Animals , Annexin A1/administration & dosage , Cell Movement/drug effects , Cytokines/metabolism , Estradiol/administration & dosage , Female , Humans , Inflammation/chemically induced , Lipopolysaccharides , Lymphocytes/drug effects , Male , Mice, Inbred C57BL , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
J Neuroendocrinol ; 24(6): 862-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22283629

ABSTRACT

The hypothalamic-pituitary-adrenocortical (HPA) responses to bacterial infection are mediated, in part, by the actions of lipopolysaccharide (LPS) on pituitary folliculostellate (FS) cells that release pro-inflammatory cytokines [e.g. interleukin (IL)-6] and thereby facilitate adrenocorticotrophic hormone (ACTH) release from neighbouring corticotrophs. In the present study, two murine pituitary cell lines [TtT/GF (FS cells) and AtT20 D16:16 (corticotrophs)], alone and in co-culture, and an in vivo model of endotoxaemia were used to examine the potential role of nuclear factor-kappa B (NF-κB) in mediating LPS-induced ACTH secretion. Both cell lines expressed mRNAs for the key components of the LPS signalling system. LPS stimulated IL-6 release from TtT/GF cells via a glucocorticoid-sensitive, NF-κB-dependent mechanism; it also activated NF-κB in AtT20 cells, as did corticotrophin-releasing hormone (CRH). IL-6 potentiated (but LPS reduced) the stimulatory effects of CRH on ACTH release from AtT20 cells, whereas blockade of NF-κB (SC-514) increased the ACTH release induced by CRH in the presence or absence of LPS. In co-cultures, CRH and LPS acted synergistically to induce release of both IL-6 and ACTH. However, although SC-514 suppressed the release of IL-6 evoked by CRH and LPS, it potentiated the concomitant increase in ACTH release. In vivo both immunological (LPS) and psychological (restraint) stress increased intrapituitary NF-κB, whereas an NF-κB inhibitor (PHA781535E) attenuated the LPS-induced release of ACTH and abolished the HPA response to restraint stress. The results obtained in the present study support the premise that NF-κB plays an important role in mediating LPS signalling in the anterior pituitary gland, particularly in relation to IL-6 and ACTH secretion, and provide novel evidence that NF-κB blockade in vivo compromises stress-induced ACTH release.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Corticotrophs/metabolism , Endotoxemia/metabolism , NF-kappa B/metabolism , NF-kappa B/physiology , Pituitary Gland, Anterior/metabolism , Animals , Cell Line , Cells, Cultured , Coculture Techniques , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Endotoxemia/pathology , Lipopolysaccharides/pharmacology , Male , Mice , Pituitary Gland, Anterior/cytology , Rats , Rats, Sprague-Dawley
4.
Inflamm Res ; 57(3): 97-103, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18369573

ABSTRACT

OBJECTIVE AND DESIGN: We have previously reported a role for annexin-A1 in liver proliferation and tumorogenicity as well as its action as an acute phase protein in a model of endotoxemia in interleukin-6 null mice. MATERIAL AND METHODS: In this study, we have investigated the analysis of the gene and protein expression in annexin-A1 null mice and the wild type livers during foetal and adult life, and in the presence of a proinflammatory stimulus. RESULTS: The data indicate a link between the expression of the annexin-A1 as serine-phosphorylated-protein during early events of the inflammatory response and as tyrosine-phosphorylated-form at later time-points, during the resolution of inflammation. CONCLUSIONS: The study of annexin-A1 post-translation modification may promote a new annexin-A1 peptide discovery programme to treat specific pathologies.


Subject(s)
Annexin A1/genetics , Endotoxemia/chemically induced , Gene Expression Regulation , Liver/physiology , Protein Processing, Post-Translational , Animals , Annexin A1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Liver/cytology , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Knockout , Promoter Regions, Genetic
5.
FASEB J ; 21(4): 1037-46, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17218541

ABSTRACT

The N-formyl peptide receptors (FPRs) are a family of G-protein coupled receptors that respond to proinflammatory N-formylated bacterial peptides (e.g., formyl-Met-Leu-Phe, fMLF) and, thus, contribute to the host response to bacterial infection. Paradoxically, a growing body of evidence suggests that some members of this receptor family may also be targets for certain anti-inflammatory molecules, including annexin A1 (ANXA1), which is an important mediator of glucocorticoid (GC) action. To explore further the potential role of FPRs in mediating ANXA1 actions, we have focused on the pituitary gland, where ANXA1 has a well-defined role as a cell-cell mediator of the inhibitory effects of GCs on the secretion of corticotrophin (ACTH), and used molecular, genetic, and pharmacological approaches to address the question in well-established rodent models. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis identified mRNAs for four FPR family members in the mouse anterior pituitary gland, Fpr-rs1, Fpr-rs2, Fpr-rs6, and Fpr-rs7. Functional studies confirmed that, like dexamethasone, ANXA1 and two ANXA1-derived peptides (ANXA1(1-188) and ANXA1(Ac2-26)) inhibit the evoked release of ACTH from rodent anterior pituitary tissue in vitro. Fpr1 gene deletion failed to modify the pituitary responses to dexamethasone or ANXA1(Ac2-26). However, lipoxin A4 (LXA4, 0.02-2 microM, a lipid mediator with high affinity for Fpr-rs1) mimicked the inhibitory effects of ANXA1 on ACTH release as also did fMLF in high (1-100 microM) but not lower (10-100 nM) concentrations. Additionally, a nonselective FPR antagonist (Boc1, 100 microM) overcame the effects of dexamethasone, ANXA1(1-188), ANXA1(Ac2-26), fMLF, and LXA4 on ACTH release, although at a lower concentration (50 microM), it was without effect. Together, the results suggest that the actions of ANXA1 in the pituitary gland are independent of Fpr1 but may involve other FPR family members, in particular, Fpr-rs1 or a closely related receptor. They thus provide the first evidence for a role of the FPR family in the regulation of neuroendocrine function.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Annexin A1/metabolism , Bacteria/metabolism , Gene Expression Regulation , Lipoxins/metabolism , Peptides/chemistry , Receptors, Formyl Peptide/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Glucocorticoids/metabolism , Male , Mice , Mice, Knockout , Pituitary Gland/metabolism , Rats , Receptors, Formyl Peptide/metabolism
6.
FASEB J ; 20(9): 1498-500, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16720734

ABSTRACT

Annexin A1 (ANXA1) has an important role in cell-cell communication in the host defense and neuroendocrine systems. In both systems, its actions are exerted extracellularly via membrane-bound receptors on adjacent sites after translocation of the protein from the cytoplasm to the cell surface of adjacent cells. This study used molecular, microscopic, and pharmacological approaches to explore the mechanisms underlying the cellular exportation of ANXA1 in TtT/GF (pituitary folliculo-stellate) cells. LPS caused serine-phosphorylation of ANXA1 (ANXA1-S27-PO4) and translocation of the phosphorylated protein to the cell membrane. The fundamental requirement of phosphorylation for membrane translocation was confirmed by immunofluorescence microscopy on cells transfected with wild-type or mutated (S27/A) ANXA1 constructs tagged with enhanced green fluorescence protein. The trafficking of ANXA1-S27-PO4 to the cell surface was dependent on PI3-kinase and MAP-kinase. It also required HMG-coenzyme A and myristoylation. The effects of HMG-coenzyme A blockade were overcome by mevalonic acid (the product of HMG-coenzyme A) and farnesyl-pyrophosphate but not by geranyl-geranylpyrophosphate or cholesterol. Together, these results suggest that serine-27 phosphorylation is essential for the translocation of ANXA1 across the cell membrane and also identify a role for isoprenyl lipids. Such lipids could target consensus sequences in ANXA1. Alternatively, they may target other proteins in the signal transduction cascade (e.g., transporters).


Subject(s)
Annexin A1/metabolism , Cell Membrane/metabolism , Protein Processing, Post-Translational , Animals , Annexin A1/genetics , Cell Communication , Cell Line, Tumor , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Genes, Reporter , Lipopolysaccharides/pharmacology , Mevalonic Acid/pharmacology , Mice , Mutagenesis, Site-Directed , Phosphorylation , Pituitary Neoplasms , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
7.
Cancer Biol Ther ; 5(6): 643-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16627980

ABSTRACT

We investigated the expression of annexin-1 (ANXA1) in thyroid carcinoma cell lines and in thyroid cancers with a different degree of differentiation. The highest level of ANXA1 expression examined by Western blotting was detected in the papillary carcinoma cells (NPA) and in the follicular cells (WRO). On the other hand, the most undifferentiated thyroid carcinoma cells (ARO and FRO) presented the lowest level of ANXA1 expression. In surgical tissue specimens from 32 patients with thyroid cancers, we found high immunoreactivity for ANXA1 in papillary (PTC) and follicular (FTC) thyroid cancers while in undifferentiated thyroid cancers (UTC) the expression of the protein was barely detectable. Control thyroid tissue resulted positive for ANXA1. In summary, 70% of UTC examined weakly expressed ANXA1, whereas 65% of PTC or FTC specimens tested showed high expression of the protein. Thus ANXA1 expression may correlate with the tumorigenesis suggesting that the protein may represent an effective differentiation marker in thyroid cancer.


Subject(s)
Annexin A1/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Cell Differentiation , Gene Expression Regulation, Neoplastic , Humans , Thyroid Gland/pathology , Tumor Cells, Cultured
9.
Biochem Soc Trans ; 32(Pt3): 507-10, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15157173

ABSTRACT

ANXA1 (annexin 1), a member of the 'annexin' family of calcium- and phospholipid-binding proteins, was originally identified as an endogenous mediator of the anti-inflammatory actions of glucocorticoids. However, this protein exerts multiple inhibitory effects on the host inflammatory response, including a preferential regulation of the adhesion step of blood-borne neutrophil within the microenvironment of an inflamed vasculature. It is now emerging that ANXA1 is endowed with other roles, since the protein is abundant in inflammatory exudates as it is produced and released by the extravasated neutrophil. In the present paper, we review the novel proapoptotic effect of ANXA1 and discuss its potential with respect to the pathophysiology of inflammation and leucocyte recruitment.


Subject(s)
Annexin A1/physiology , Apoptosis , Neutrophils/pathology , Animals , Annexin A1/metabolism , Humans , Inflammation , Leukocytes/pathology
10.
J Neuroendocrinol ; 15(12): 1134-43, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14636175

ABSTRACT

Annexin 1 (ANXA1) is a key mediator of the inhibitory effects of glucocorticoids on adrenocorticotropic hormone (ACTH) release, which develop within 1-2 h of a steroid challenge. Our previous studies, which showed that (i) ANXA1 is expressed principally by the nonsecretory folliculo-stellate cells in the pituitary gland; (ii) glucocorticoids cause the exportation of ANXA1 from these cells; and (iii) corticotrophs express specific ANXA1 binding sites, led us to propose that ANXA1 serves as a paracrine or juxtacrine mediator of glucocorticoids. To address this hypothesis, we examined ANXA1-dependent glucocorticoid actions in co-cultures of murine corticotroph (AtT20 clone D1) and folliculo-stellate (TtT/GF) cell lines. ANXA1 mRNA and protein were found in abundance in TtT/GF cells but neither was detectable in the AtT20 cells. AtT20 cells (alone and in co-culture with TtT/GF cells) responded to corticotropin-releasing hormone (CRH) (0.1-1 micro m) with increased ACTH release. The CRH-stimulated release of ACTH from AtT20 cells cultured alone was unaffected by preincubation with dexamethasone (Dex, 100 nm); by contrast, in co-cultures of AtT20 and TtT/GF cells, the steroid readily inhibited the secretory response to CRH. The effects of Dex on ACTH release were mimicked by N-terminal ANXA1 fragments (ANXA1Ac2-26, 2 micro g/ml and ANXA11-188, 0.1 ng/ml) and reversed by mifepristone (1 micro m) and by an antisense oligodeoxynucleotide (ODN) to ANXA1 (50 nm) but not by control ODNs. The antisense ODN also specifically blocked the Dex-induced externalization of ANXA1 from TtT/GF cells. Immunofluorescence imaging of the co-cultures localized the exported protein to the vicinity of the AtT20 cells and identified ANXA1 binding sites on these cells. These results provide functional and histological evidence to support our premise that the early inhibitory effects of glucocorticoids on ACTH release are dependent upon paracrine/juxtacrine actions of ANXA1 derived from folliculo-stellate cells.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Annexin A1/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Pituitary Gland/cytology , Pituitary Gland/metabolism , Pituitary Neoplasms , Animals , Annexin A1/genetics , Cell Line, Tumor , Coculture Techniques , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Paracrine Communication/drug effects , Paracrine Communication/physiology , RNA, Messenger/analysis
11.
J Neuroendocrinol ; 15(10): 946-57, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12969239

ABSTRACT

Our previous studies have identified a role for annexin 1 (ANXA1), a protein produced by the pituitary folliculostellate cells, as a paracrine/juxtacrine mediator of the acute regulatory effects of glucocorticoids on the release of adrenocorticotropic hormone and other pituitary hormones. In the present study, we focused on the secretion of thyroid stimulating hormone (TSH) and luteinizing hormone (LH) and used a battery of ANXA1-derived peptides to identify the key domains in the ANXA1 molecule that are critical to the inhibition of peptide release. In addition, as ANXA1 is a substrate for protein kinase C (PKC) and tyrosine kinase, we examined the roles of these kinases in the manifestation of the ANXA1-dependent inhibitory actions of dexamethasone on TSH and LH release. Dexamethasone suppressed the forskolin-induced release of TSH and LH from rat anterior pituitary tissue in vitro. Its effects were mimicked by human recombinant ANXA1 (hrANXA1) and a truncated protein, ANXA1(1-188). ANXA1(Ac2-26), also suppressed stimulated peptide release but it lacked both the potency and the efficacy of the parent protein. Shorter N-terminal ANXA1 sequences were without effect. The PKC inhibitor PKC(19-36) abolished the inhibitory actions of dexamethasone on the forskolin-evoked release of TSH and LH; it also attenuated the inhibitory actions of ANXA1(Ac2-26). Similar effects were produced by annexin 5 (ANXA5) which sequesters PKC in other systems. By contrast, the tyrosine kinase inhibitors, p60v-src (137-157) and genistein, had no effect on the secretion of TSH or LH alone or in the presence of forskolin and/or dexamethasone. Dexamethasone caused the translocation of a tyrosine-phosphorylated species of ANXA1 to the surface of pituitary cells. The total amount of ANXA1 exported from the cells in response to the steroid was unaffected by tyrosine kinase blockade. However, the degree of tyrosine-phosphorylation of the exported protein was markedly reduced by genistein. These results suggest that (i) the ANXA1-dependent inhibitory actions of dexamethasone on the release of TSH and LH require PKC and sequences in the N-terminal domain of ANXA1, but are independent of tyrosine kinase, and (ii) while dexamethasone induces the cellular exportation of a tyrosine-phosphorylated species of ANXA1, tyrosine phosphorylation per se is not critical to the steroid-induced passage of ANXA1 across the membrane.


Subject(s)
Annexin A1/pharmacology , Glucocorticoids/pharmacology , Luteinizing Hormone/metabolism , Phosphotransferases/physiology , Thyrotropin/metabolism , Amino Acid Sequence , Animals , Annexin A5/pharmacology , Blotting, Western , Colforsin/pharmacology , Electrophoresis, Polyacrylamide Gel , In Vitro Techniques , Male , Membrane Proteins/chemistry , Molecular Sequence Data , Peptides/pharmacology , Phosphorylation , Phosphotransferases/antagonists & inhibitors , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley
12.
Am J Pathol ; 159(4): 1435-43, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583971

ABSTRACT

Annexin 1 (ANXA1) is a calcium-binding protein endowed with anti-inflammatory properties. Using an extra-hepatic system, we showed that interleukin (IL)-6 regulates ANXA1 expression at the transcriptional level. The purpose of this study was to determine whether ANXA1 synthesis was modulated by IL-6 during experimental inflammation. We have compared liver ANXA1 expression during systemic and localized inflammatory reaction, using lipopolysaccharide (LPS) and turpentine. LPS treatment strongly induced ANXA1 expression in the liver of wild-type (WT) animals (+600%) whereas a modest increase (+60%) was measured in IL-6 knockout (KO) animals. Turpentine treatment did not affect the expression of ANXA1 in either animal type. LPS enhanced serum corticosteroid levels equally in WT and IL-6 KO mice, whereas higher tumor necrosis factor (TNF)-alpha and IL-1beta levels were released in IL-6 KO animals. Injection of mouse recombinant IL-6 to IL-6 KO animals before LPS or TNF-alpha challenge, replenished ANXA1 liver synthesis to that of WT animals. Exogenous ANXA1 but not ANXA5, administered to IL-6 KO mice before LPS challenge inhibited TNF-alpha release. We propose that ANXA1 acts as a novel acute phase protein, which is controlled in the liver by TNF-alpha and IL-6, and which may contribute to the resolution of systemic endotoxemia through a negative feedback on TNF-alpha release.


Subject(s)
Annexins/metabolism , Cytokines/physiology , Endotoxemia/metabolism , Liver/metabolism , Animals , Antibodies/pharmacology , Corticosterone/blood , Hepatocytes/metabolism , Immunohistochemistry , Injections , Interleukin-1/antagonists & inhibitors , Interleukin-1/blood , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Osmolar Concentration , Recombinant Proteins/pharmacology , Reference Values , Tissue Distribution , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
13.
Clin Exp Allergy ; 31(7): 1116-25, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11468004

ABSTRACT

BACKGROUND: Annexin-1 (ANXA1, lipocortin 1) is a pleiotrophic protein produced by many cell types including peripheral blood leucocytes. Although it has been shown to inhibit "macroscopic" inflammatory processes in animal models, its direct effects on antigen-activated human T cells have not been studied. OBJECTIVE: To test the hypothesis that ANXA1-derived peptides inhibit antigen-driven prototype Th1 and Th2-type human T cell responses of clinical relevance and lectin-driven responses in vitro. METHODS: Peripheral blood mononuclear cells (PBMC) were isolated from 14 atopic subjects sensitized to house dust mite allergen (Dermatophagoides pteronyssinus, Der p) and purified protein derivative (PPD) of Mycobacterium tuberculosis. PBMC (1 x 106/mL) were cultured with phytohaemagglutinin (PHA; 5 microg/mL; 4 days), Der p (25 microg/mL; 6 days), PPD (10 microg/mL, 6 days) or medium control. Two ANXA1-derived peptides, Ac2-26 and AF-2 (5-500 microM), were assessed for possible inhibition of PHA-and antigen-induced T cell proliferation (measured by 3H-thymidine uptake), while Ac2-26 was assessed for inhibition of Der p-induced interleukin (IL)-5 release and PPD-induced interferon-gamma (IFN-gamma) release (measured by ELISA). Comparison was made with dexamethasone as an established inhibitory control. Endogenous production by PBMC of cell surface-associated and intracellular ANXA1 in response to PHA, Der p and PPD in the presence and absence of dexamethasone was measured by specific ELISA. RESULTS: Both PHA- and antigen-induced T cellular proliferation were inhibited by dexamethasone. Although neither ANXA1-derived peptide significantly altered PHA-induced proliferation, both effected concentration-dependent reductions in antigen-induced proliferation, Ac2-26 being the more potent. Peptides of identical amino acid composition to Ac2-26 and AF-2, but of random sequence, were ineffective at equivalent concentrations. In addition, Ac2-26 and dexamethasone inhibited Der p-induced IL-5 release and PPD-induced IFN-gamma release in a concentration-dependent fashion. Endogenous ANXA1 was detectable in PBMC, but at concentrations approximately 104-fold lower, in molar terms, than the effective concentrations of the exogenously added, ANXA1-derived inhibitory peptides. Endogenous production was not significantly altered by any of the T cell stimuli employed in this study, in the presence or absence of dexamethasone. CONCLUSION: In prototype Th1 and Th2-type human T cell responses, ANXA1-derived peptides can inhibit antigen-driven cellular proliferation and cytokine production.


Subject(s)
Annexin A1/pharmacology , Peptides/pharmacology , Th1 Cells/immunology , Th2 Cells/immunology , Adult , Amino Acid Sequence , Antigens/immunology , Antigens, Dermatophagoides , Antigens, Surface/biosynthesis , Cell Division/drug effects , Cells, Cultured , Dexamethasone/pharmacology , Female , Glycoproteins/immunology , Humans , Lectins/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Male , Middle Aged , Molecular Sequence Data , Phytohemagglutinins/immunology , Th1 Cells/drug effects , Th2 Cells/drug effects , Tuberculin/immunology
14.
Am J Pathol ; 158(6): 1969-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395373

ABSTRACT

An innovative avenue for anti-inflammatory therapy is inhibition of neutrophil extravasation by potentiating the action of endogenous anti-inflammatory mediators. The glucocorticoid-inducible protein annexin 1 and derived peptides are effective in inhibiting neutrophil extravasation. Here we tested the hypothesis that an interaction with the receptor for formylated peptide (FPR), so far reported only in vitro, could be the mechanism for this in vivo action. In a model of mouse peritonitis, FPR antagonists abrogated the anti-migratory effects of peptides Ac2-26 and Ac2-12, with a partial reduction in annexin 1 effects. A similar result was obtained in FPR (knock-out) KO mice. Binding of annexin 1 to circulating leukocytes was reduced (>50%) in FPR KO mice. In vitro, annexin binding to peritoneal macrophages was also markedly reduced in FPR KO mice. Finally, evidence of direct annexin 1 binding to murine FPR was obtained with HEK-293 cells transfected with the receptor. Overall, these results indicate a functional role for FPR in the anti-migratory effect of annexin 1 and derived peptides.


Subject(s)
Annexin A1/pharmacology , Chemotaxis, Leukocyte , Peptide Fragments/pharmacology , Receptors, Immunologic/physiology , Receptors, Peptide/physiology , Animals , Annexin A1/metabolism , Cell Line , Leukocytes/metabolism , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Peptides , Peritonitis/immunology , Receptors, Formyl Peptide , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/genetics
15.
Br J Pharmacol ; 133(2): 217-28, 2001 May.
Article in English | MEDLINE | ID: mdl-11350857

ABSTRACT

Transfection of the pre-monomyelocytic U937 cell line with a plasmid coding for full-length annexin 1 (ANX1, 347 amino acid) leads to cell death by promoting apoptosis. In addition, over-expression of the N-terminal and the first domain of the protein (144 amino acids, clone ANX1-S), which does not contain the Ca2+ binding sites, gives susceptibility to cell apoptosis following activation by either 5 ng ml(-1) tumour necrosis factor (TNF)-alpha or 1 - 40 microg ml-1 etoposide. This was demonstrated by using the fluorescent labelled annexin V, cell cycle and nuclear staining analyses. Transfection with an empty plasmid (clone CMV) or with a plasmid carrying the cDNA antisense for ANX1 (clone ANX1-AS) did not alter U937 cells to the degree of apoptosis promoted by either stimulant. Treatment of CMV U937 cells with TNF-alpha increased ANX1 mRNA and protein expression in a time-dependent manner, with maximal increases at 3 and 6 h, respectively. Clone ANX1-S showed higher constitutive (more than 2 fold) and activated caspase-3 activity, associated with higher phospholipase A2 (PLA2) activity (in the region of +50 - 100%), whereas expression of cytosolic PLA2 Bax and Bcl-2 were similar in all cell clones, as determined by Western blotting. In conclusion, this study demonstrates a complex regulatory role of cell apoptosis for ANX1, at least with regards to cells of the myelo-monocytic lineage.


Subject(s)
Annexin A1/genetics , Apoptosis/genetics , Apoptosis/physiology , Caspases/physiology , Monocytes/metabolism , Arachidonic Acid/metabolism , Benzimidazoles , Binding Sites/genetics , Calcium/metabolism , Caspase 3 , Cell Adhesion Molecules/metabolism , Cell Cycle/physiology , Enzyme Activation/physiology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Monocytes/enzymology , Mutagenesis, Site-Directed , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
16.
FASEB J ; 14(13): 1867-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023969

ABSTRACT

We assessed here the effect of the glucocorticoid-regulated protein lipocortin 1 (LC1) in a model of rat myocardial ischemia reperfusion. Treatment of animals with human recombinant LC1 at the end of a 25-min ischemic period significantly reduced the extent of infarct size in the area at risk as measured 2 h later, with approximately 50% inhibition at the highest dose tested of 50 microg per rat (equivalent to 5.4 nmol/kg). The protective effect of LC1 was abolished by protein denaturation and not mimicked by the structurally related protein annexin V. A combination of electron and light microscopy techniques demonstrated the occurrence of the myocardial damage at the end of the reperfusion period, with loss of fiber organization. LC1 provided a partial and visible protection. The dose-dependent protection afforded by LC1 was paralleled by lower values of myeloperoxidase activity, tumor necrosis factor a, and macrophage inflammatory protein-1a. The functional link between migrated leukocytes and the myocardial damage was confirmed by electron and light microscopy, and a significantly lower number of extravasated leukocytes was counted in the group of rats treated with LC1 (50 microg). In conclusion, we demonstrate for the first time that LC1 reduces the leukocyte-dependent myocardial damage associated with an ischemia-reperfusion procedure.


Subject(s)
Annexin A1/therapeutic use , Chemotaxis, Leukocyte , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Animals , Cell Adhesion/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Models, Biological , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Neutrophils , Rats , Recombinant Proteins/therapeutic use
17.
J Immunol ; 165(3): 1573-81, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10903766

ABSTRACT

Annexin 1 (ANX1), a calcium-binding protein, participates in the regulation of early inflammatory responses. Whereas some of its effects depend on intracellular interactions, a growing number of observations indicate that ANX1 may also act via autocrine/paracrine functions following externalization to the outer side of the plasma membrane. We studied the effects of ANX1 on leukocyte adhesion to endothelial cells using as a model system the monocytic cell line U937 and human bone marrow microvascular endothelial cells. Exogenous rANX1, as well as endogenous ANX1 externalized by U937 differentiated in vitro, inhibited monocyte firm adhesion to vascular endothelium. Both binding of ANX1 to U937 cells and ANX1-mediated inhibition of cell adhesion involved the short N-terminal domain of the ANX1 molecule. Under experimental conditions in which ANX1 inhibited U937 adhesion to human bone marrow microvascular endothelial cells, this protein specifically colocalized with the alpha 4 integrin, and a direct interaction between ANX1 and the alpha 4 integrin could be documented by immunoprecipitation experiments. Moreover, ANX1 competed with the endothelial integrin counterreceptor, VCAM-1, for binding to alpha 4 integrin. These results indicate that ANX1 plays an important physiological role in modulating monocyte firm adhesion to the endothelium.


Subject(s)
Annexin A1/immunology , Annexin A1/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Integrins/physiology , Monocytes/immunology , Monocytes/metabolism , Receptors, Lymphocyte Homing/physiology , Autocrine Communication/immunology , Binding Sites/immunology , Binding, Competitive/immunology , Cell Adhesion/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Integrin alpha4beta1 , Integrins/metabolism , Ligands , Lymphocyte Function-Associated Antigen-1/metabolism , Microcirculation/cytology , Microcirculation/immunology , Microcirculation/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Receptors, Lymphocyte Homing/metabolism , U937 Cells , Vascular Cell Adhesion Molecule-1/metabolism
18.
Life Sci ; 66(18): PL265-70, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10809174

ABSTRACT

The potential involvement of endogenous lipocortin 1 in the process of cellular apoptosis, particularly in cells of the myelo-monocytic lineage, has been investigated. U937 cells were transfected either with an antisense or a sense DNA for lipocortin 1 and the stable clones 36.4AS clone (20-40% lower lipocortin 1 levels) and 15S (30% higher lipocortin 1 levels) were obtained. Cell apoptosis was induced by incubation with tumor necrosis factor-alpha: optimal responses were observed within a 24 h incubation period at a 5 ng/ml concentration. Apoptosis was assessed both morphologically, by annexin V binding and cell cycle analysis with propidium iodide. Whilst no consistent difference was seen between wild type cells and clone 36.4AS, a higher incidence of apoptosis (ranging from +30% to + 60%) was observed in the 15S clone. Release of arachidonic acid from loaded cells was promoted by 24 h incubation with the cytokine, and a higher degree of release was measured in the 15S clone. These data indicate that endogenous intracellular lipocortin 1 is involved in the promotion of apoptosis in cells of the myelo-monocytic derivation.


Subject(s)
Annexin A1/biosynthesis , Apoptosis/physiology , Apoptosis/drug effects , Arachidonic Acid/metabolism , Cell Cycle/physiology , Clone Cells/drug effects , Clone Cells/metabolism , Flow Cytometry , Humans , Phospholipases A/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
19.
J Cell Physiol ; 183(1): 65-73, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10699967

ABSTRACT

We studied the long-terms effects of interleukin-1beta (IL-1beta; 3 to 6 h) on alpha-(methylamino) isobutyric acid (MeAIB), a nonmetabolizable amino acid transported by system A. We found that IL-1beta induced a large decrease in MeAIB uptake by human osteoarthritic synovial cells and a concomitant increase in prostaglandin E(2) (PGE(2)) synthesis. Therefore, we investigated whether PGE(2) acts as a mediator for the long-term action of IL-1beta. We found that exogenous PGE(2) inhibited MeAIB uptake, and that AH6809, a PGE(2) receptor antagonist, inhibited IL-1beta-mediated MeAIB uptake. To identify the enzymes involved in the IL-1beta-mediated synthesis of PGE(2) that inhibits MeAIB uptake, we studied the expression of secreted (s) and cytosolic (c) phospholipase A(2) (PLA(2)). Because both were expressed, we selected a broad spectrum of inhibitors to determine which of the two PLA(2)s was involved. We used AACOCF3, a cPLA(2) inhibitor, and dithiothreitol (DTT) and bromophenacyl bromide (BPB), which are sPLA(2) inhibitors. Our results suggest that the PLA(2) involved in the IL-1beta-mediated synthesis of PGE(2) was sPLA(2). We also showed the expression of cyclooxygenase (COX)-2 and its partial involvement using a potent selective COX-2 inhibitor, L-745337. These findings provide insight into the mechanisms underlying the IL-1beta-mediated regulation of transport system A. The Il-1beta-induced inhibition of MeAIB uptake in human osteoarthritic synovial cells thus seems to be essentially mediated by PGE(2) production via the activation of sPLA(2) and the partial activation of COX-2.


Subject(s)
Carrier Proteins/metabolism , Dinoprostone/physiology , Interleukin-1/pharmacology , Osteoarthritis/physiopathology , Second Messenger Systems/physiology , Synovial Membrane/physiopathology , Xanthones , beta-Alanine/analogs & derivatives , Amino Acid Transport Systems , Arachidonic Acids/pharmacology , Biological Transport/drug effects , Carrier Proteins/drug effects , Cells, Cultured , Cyclooxygenase 2 , Cytosol/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Indans/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Membrane Proteins , Osteoarthritis/pathology , Phospholipases A/genetics , Phospholipases A/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Second Messenger Systems/drug effects , Synovial Membrane/drug effects , Synovial Membrane/pathology , Xanthenes/pharmacology , beta-Alanine/pharmacokinetics
20.
Hepatology ; 31(2): 371-80, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10655260

ABSTRACT

We have used a transgenic animal model, which constitutively develops hepatocarcinoma (Antithrombin III SV40 T large Antigen: ASV), to study the involvement of Annexin 1 (ANX1) in liver regeneration and malignant transformation. Primary hepatocytes isolated from normal mice did not express ANX1. In contrast, ANX1 was strongly expressed in hepatocytes of transgenic mice during constitutive development of hepatocarcinoma. In ASV transgenic mice, an elevated ANX1 level preceded the appearance of the tumor, indicating that it could be a good marker in the diagnosis of cancer. One-third hepatectomy in normal mice resulted in stimulation of ANX1 synthesis and phosphorylation. This upregulation correlated with increased synthesis of EGF and consequently with increased phosphorylation of the EGF receptor (EGF-R). Stable transfection of a hepatocyte cell line derived from ASV transgenic mice (mhAT2) with antisense complementary DNA for ANX1 reduced the proliferation rate as well as cytosolic phospholipase A(2) (cPLA(2)) activity. Thus, ANX1 expression and phosphorylation could be a factor implicated in liver regeneration and tumorigenesis, either through modulation of cPLA(2) activity or EGF-R function.


Subject(s)
Annexin A1/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic , Liver Neoplasms/metabolism , Liver Regeneration/physiology , Animals , Antigens, Polyomavirus Transforming/genetics , Antithrombin III/genetics , Cells, Cultured , Epidermal Growth Factor/pharmacology , Hepatectomy/methods , Mice , Mice, Inbred Strains , Mice, Transgenic/genetics , Phosphorylation/drug effects , Postoperative Period , Promoter Regions, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...