Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Molecules ; 26(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34946686

ABSTRACT

Glioblastoma is an aggressive cancer, against which medical professionals are still quite helpless, due to its resistance to current treatments. Scorpion toxins have been proposed as a promising alternative for the development of effective targeted glioblastoma therapy and diagnostic. However, the exploitation of the long peptides could present disadvantages. In this work, we identified and synthetized AaTs-1, the first tetrapeptide from Androctonus australis scorpion venom (Aa), which exhibited an antiproliferative effect specifically against human glioblastoma cells. Both the native and synthetic AaTs-1 were endowed with the same inhibiting effect on the proliferation of U87 cells with an IC50 of 0.56 mM. Interestingly, AaTs-1 was about two times more active than the anti-glioblastoma conventional chemotherapeutic drug, temozolomide (TMZ), and enhanced its efficacy on U87 cells. AaTs-1 showed a significant similarity with the synthetic peptide WKYMVm, an agonist of a G-coupled formyl-peptide receptor, FPRL-1, known to be involved in the proliferation of glioma cells. Interestingly, the tetrapeptide triggered the dephosphorylation of ERK, p38, and JNK kinases. It also enhanced the expression of p53 and FPRL-1, likely leading to the inhibition of the store operated calcium entry. Overall, our work uncovered AaTs-1 as a first natural potential FPRL-1 antagonist, which could be proposed as a promising target to develop new generation of innovative molecules used alone or in combination with TMZ to improve glioblastoma treatment response. Its chemical synthesis in non-limiting quantity represents a valuable advantage to design and develop low-cost active analogues to treat glioblastoma cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma , Oligopeptides/pharmacology , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Scorpion Venoms/chemistry , Tumor Suppressor Protein p53/biosynthesis , Up-Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Oligopeptides/chemistry , Scorpions
2.
Cells ; 9(4)2020 04 10.
Article in English | MEDLINE | ID: mdl-32290034

ABSTRACT

We reported earlier that an anti-inflammatory small peptide receptor-formyl peptide receptor-2 (FPR2) was significantly decreased in placentas from third trimester pregnancies complicated with fetal growth restriction (FGR), compared to placentas from uncomplicated control pregnancies, suggesting FPR2 may play a role in the development of FGR. The aim of this study is to investigate whether the actions of FPR2 alters placental growth process in humans. Accordingly, using small-for-gestation age (SGA) as a proxy for FGR, we hypothesize that FPR2 expression is decreased in first-trimester placentas of women who later manifest FGR, and contributes to aberrant trophoblast function and the development of FGR. Chorionic villus sampling (CVS) tissues were collected at 10-12 weeks gestation in 70 patients with singleton fetuses; surplus tissue was used. Real-time PCR and immunoassays were performed to quantitate FPR2 gene and protein expression. Silencing of FPR2 was performed in two independent, trophoblast-derived cell lines, HTR-8/SVneo and JEG-3 to investigate the functional consequences of FPR2 gene downregulation. FPR2 mRNA relative to 18S rRNA was significantly decreased in placentae from SGA-pregnancies (n = 28) compared with controls (n = 52) (p < 0.0001). Placental FPR2 protein was significantly decreased in SGA compared with control (n = 10 in each group, p < 0.05). Proliferative, migratory and invasive potential of the human placental-derived cell lines, HTR-8/SVneo and JEG-3 were significantly reduced in siFPR2 treated cells compared with siCONT control groups. Down-stream signaling molecules, STAT5B and SOCS3 were identified as target genes of FPR2 action in the trophoblast-derived cell lines and in SGA and control chorionic villous tissues. FPR2 is a novel regulator of key molecular pathways and functions in placental development, and its decreased expression in women destined to develop FGR reinforces a placental origin of SGA/FGR, and that it contributes to causing the development of SGA/FGR.


Subject(s)
Infant, Small for Gestational Age , Placenta/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Adult , Epithelial-Mesenchymal Transition , Female , Humans , Infant, Newborn , Pregnancy , Pregnancy Trimester, First , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Signal Transduction
3.
Ann Diagn Pathol ; 37: 62-66, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30286327

ABSTRACT

Astrocytomas represent the majority of cerebral gliomas. Studies show that the anti-inflammatory protein Annexin-A1 (ANXA1) is associated with the tumor invasion process and that its actions can be mediated by the receptor for formylated peptides (FPR). Therefore, we evaluated the expression of ANXA1, the receptor FPR2 and matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) in brain astrocytomas. Detection of proteins was performed in sections of diffuse astrocytomas (grade II), anaplastic astrocytomas (grade III) and glioblastomas (GBM, grade IV) and quantifications were made by densitometry. Our analyses showed increased expression of ANXA1 in astrocytomas of all grades, but especially in GBM. The expression of FPR2 is similar to that found for ANXA1, being higher in GBM. Immunostaining for MMPs is also stronger as the degree of malignancy increases, especially with respect to MMP-9. The positive correlation between ANXA1/FPR2 and ANXA1/MMP-9 was observed in all tumors studied. The data indicate the possible action of ANXA1 and FPR2 on the development and progression of astrocytomas, related to increased expression of MMP-9. Thereby, ANXA1 and FPR2 are involved in the biology and malignancy of diffuse astrocytic tumors.


Subject(s)
Annexin A1/biosynthesis , Astrocytoma/pathology , Biomarkers, Tumor/biosynthesis , Brain Neoplasms/pathology , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Adult , Aged , Female , Humans , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Middle Aged
4.
Neurochem Res ; 43(8): 1587-1598, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948727

ABSTRACT

Arachidonic acid and docosahexaenoic acid (DHA) released by the action of phospholipases A2 (PLA2) on membrane phospholipids may be metabolized by lipoxygenases to the anti-inflammatory mediators lipoxin A4 (LXA4) and resolvin D1 (RvD1), and these can bind to a common receptor, formyl-peptide receptor 2 (FPR2). The contribution of this receptor to axonal or dendritic outgrowth is unknown. The present study was carried out to elucidate the distribution of FPR2 in the rat CNS and its role in outgrowth of neuronal processes. FPR2 mRNA expression was greatest in the brainstem, followed by the spinal cord, thalamus/hypothalamus, cerebral neocortex, hippocampus, cerebellum and striatum. The brainstem and spinal cord also contained high levels of FPR2 protein. The cerebral neocortex was moderately immunolabelled for FPR2, with staining mostly present as puncta in the neuropil. Dentate granule neurons and their axons (mossy fibres) in the hippocampus were very densely labelled. The cerebellar cortex was lightly stained, but the deep cerebellar nuclei, inferior olivary nucleus, vestibular nuclei, spinal trigeminal nucleus and dorsal horn of the spinal cord were densely labelled. Electron microscopy of the prefrontal cortex showed FPR2 immunolabel mostly in immature axon terminals or 'pre-terminals', that did not form synapses with dendrites. Treatment of primary hippocampal neurons with the FPR2 inhibitors, PBP10 or WRW4, resulted in reduced lengths of axons and dendrites. The CNS distribution of FPR2 suggests important functions in learning and memory, balance and nociception. This might be due to an effect of FPR2 in mediating arachidonic acid/LXA4 or DHA/RvD1-induced axonal or dendritic outgrowth.


Subject(s)
Axons/metabolism , Brain/metabolism , Dendrites/metabolism , Receptors, Lipoxin/biosynthesis , Spinal Cord/metabolism , Animals , Axons/chemistry , Axons/ultrastructure , Brain/ultrastructure , Brain Chemistry/physiology , Cell Survival/physiology , Central Nervous System/chemistry , Central Nervous System/metabolism , Central Nervous System/ultrastructure , Dendrites/chemistry , Dendrites/ultrastructure , Male , Rats , Rats, Wistar , Receptors, Lipoxin/analysis , Spinal Cord/chemistry , Spinal Cord/ultrastructure
5.
J Immunol ; 195(6): 2852-60, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26276873

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a common, progressive, and invariably lethal interstitial lung disease with no effective therapy. The key cell driving the development of fibrosis is the myofibroblast. Lipoxin A4 (LXA4) is an anti-inflammatory lipid, important in the resolution of inflammation, and it has potential antifibrotic activity. However, the effects of LXA4 on primary human lung myofibroblasts (HLMFs) have not previously been investigated. Therefore, the aim of this study was to examine the effects of LXA4 on TGF-ß1-dependent responses in IPF- and nonfibrotic control (NFC)-derived HLMFs. HLMFs were isolated from IPF and NFC patients and grown in vitro. The effects of LXA4 on HLMF proliferation, collagen secretion, α-smooth muscle actin (αSMA) expression, and Smad2/3 activation were examined constitutively and following TGF-ß1 stimulation. The LXA4 receptor (ALXR) was expressed in both NFC- and IPF-derived HLMFs. LXA4 (10(-10) and 10(-8) mol) reduced constitutive αSMA expression, actin stress fiber formation, contraction, and nuclear Smad2/3, indicating regression from a myofibroblast to fibroblast phenotype. LXA4 also significantly inhibited FBS-dependent proliferation and TGF-ß1-dependent collagen secretion, αSMA expression, and Smad2/3 nuclear translocation in IPF-derived HLMFs. LXA4 did not inhibit Smad2/3 phosphorylation. In summary, LXA4 attenuated profibrotic HLMF activity and promoted HLMF regression to a quiescent fibroblast phenotype. LXA4 or its stable analogs delivered by aerosol may offer a novel approach to the treatment of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/pathology , Lipoxins/pharmacology , Myofibroblasts/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Transforming Growth Factor beta1/pharmacology , Actins/biosynthesis , Cell Proliferation , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Enzyme Activation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/immunology , Inflammation/immunology , Inflammation/pathology , Lung/cytology , Lung/pathology , Phosphorylation/drug effects , RNA, Messenger/biosynthesis , Smad2 Protein/metabolism , Smad3 Protein/metabolism
6.
J Immunol ; 194(11): 5161-73, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917089

ABSTRACT

Systemic sclerosis (SSc) is characterized by chronic inflammation and fibrosis. N-Formyl peptide (fMLF) receptors (FPRs) are chemotactic receptors involved in inflammation. Three FPRs have been identified: FPR1, FPR2, and FPR3. We have examined, by RT-PCR, Western blot and immunohistochemistry, FPRs expression in skin fibroblasts from 10 normal subjects and 10 SSc patients, showing increased expression in SSc fibroblasts. Several functions of FPRs occur through the interaction with a region of the urokinase-type plasminogen activator receptor (uPAR88-92), able to interact with FPRs and to mediate urokinase (uPA) or fMLF-dependent cell migration. Soluble uPAR84-95 peptide can act as a direct ligand of FPRs. Furthermore, uPA or its aminoterminal fragment (ATF) can promote the exposure of the uPAR88-92 region. The WKYMVm peptide is a FPRs pan-agonist. We investigated the functional effects of these agonists on normal and SSc fibroblasts. ATF, uPAR84-95, and WKYMVm regulated adhesion, migration, and proliferation of normal fibroblasts. Despite FPR overexpression, the response of SSc fibroblasts to the same agonists was greatly reduced, except for the proliferative response to ATF. SSc fibroblasts showed increased α-smooth muscle actin expression and improved capability to induce wound closure. Indeed, they overexpressed a cleaved uPAR form, exposing the uPAR88-92 region, and vitronectin, both involved in fibrosis and in the fibroblast-to-myofibroblast transition. FPR stimulation promoted α-smooth muscle actin expression in normal fibroblasts as well as motility, matrix deposition, αvß5 integrin expression, and radical oxygen species generation in normal and SSc fibroblasts. This study provides evidence that FPRs may play a role in fibrosis and in the fibroblast-to-myofibroblast transition.


Subject(s)
Fibrosis/pathology , Myofibroblasts/cytology , Receptors, Formyl Peptide/metabolism , Scleroderma, Systemic/pathology , Actins/biosynthesis , Adult , Aged , Cell Adhesion/drug effects , Cell Differentiation , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibrosis/immunology , Humans , Inflammation/immunology , Male , Middle Aged , Myofibroblasts/metabolism , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Receptors, Vitronectin/biosynthesis , Scleroderma, Systemic/immunology , Skin/metabolism , Transcriptional Activation , Urokinase-Type Plasminogen Activator/metabolism , Vitronectin , Wound Healing/physiology
7.
Am J Physiol Cell Physiol ; 306(2): C178-85, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24259417

ABSTRACT

Sjögren's syndrome (SS) is an autoimmune disorder characterized by chronic inflammation and destruction of salivary and lacrimal glands, leading to dry mouth, dry eyes, and the presence of anti-nuclear antibodies. Despite modern advances, the current therapies for SS have no permanent benefit. A potential treatment could involve the use of resolvins, which are highly potent endogenous lipid mediators that are synthesized during the resolution of inflammation to restore tissue homeostasis. Our previous studies indicate that ALX/FPR2, the receptor for RvD1, is expressed and active in the rat parotid cell line Par-C10. Specifically, activation of ALX/FPR2 with RvD1 blocked inflammatory signals caused by TNF-α and enhanced salivary epithelial integrity. The goal of this study was to investigate RvD1 receptor expression and signaling pathways in primary salivary cells. Additionally, we determined the role of the aspirin-triggered 17R analog (AT-RvD1, a more chemically stable RvD1 epimeric form) in prevention of TNF-α-mediated salivary inflammation in mouse submandibular glands (mSMG). Our results indicate that ALX/FPR2 is expressed in mSMG and is able to elicit intracellular Ca2+ responses and phosphorylation of Erk1/2, as well as Akt. Given that these signaling pathways are linked to cell survival, we investigated whether AT-RvD1 was able to prevent programmed cell death in mSMG. Specifically, we determined that AT-RvD1 prevented TNF-α-mediated caspase-3 activation. Finally, we show that ALX/FPR2 is expressed in human minor salivary glands with and without SS, indicating the potential therapeutic use of AT-RvD1 for this condition.


Subject(s)
Docosahexaenoic Acids/biosynthesis , Gene Expression Regulation , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Salivary Glands/physiology , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Salivary Glands/pathology , Signal Transduction/physiology , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology
8.
Ann Otol Rhinol Laryngol ; 122(11): 683-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24358628

ABSTRACT

OBJECTIVES: The resolution of inflammation is an active process controlled by several anti-inflammatory and pro-resolution mediators. Lipoxin A4, an endogenous lipid mediator, is a potential pro-resolution mediator that could attenuate inflammation. This study was conducted to elucidate the role of lipoxin A4 in upper airway inflammation. METHODS: Nasal secretions were collected from patients with chronic rhinosinusitis with nasal polyposis, patients with allergic rhinitis, and control subjects. The concentration of lipoxin A4 was measured by enzyme-linked immunosorbent assay. Nasal tissues were obtained from nasal polyps and inferior turbinates during endonasal surgery. The mRNA expressions of lipoxygenases (LOXs), lipoxin receptor (formyl peptide receptor-like 1; FPRL-1), and cysteinyl leukotriene type 1 receptor (CysLT1R) in nasal tissues were examined by reverse-transcription polymerase chain reaction. Tissue localization of FPRL-1 was determined by immunohistochemical staining. The in vitro effect of lipoxin A4 on airway epithelial cells was also examined. RESULTS: A significant concentration of lipoxin A4 was found in nasal secretions, and the concentration was increased in patients with allergic rhinitis. The mRNA expressions of 5-LOX, 15-LOX-1, FPRL-1, and CysLT1R were significantly greater in nasal polyps than in inferior turbinates. FPRL-1 was localized in nasal epithelial cells. Lipoxin A4 inhibited tumor necrosis factor alpha-induced interleukin 8 release from airway epithelial cells via its receptor FPRL-1. CONCLUSIONS: These results indicate that lipoxin A4 may play a role in the resolution of upper airway inflammation. A low concentration of lipoxin A4 may be involved in chronic inflammation of the upper airways.


Subject(s)
Gene Expression Regulation , Lipoxins/genetics , Nasal Polyps/genetics , RNA, Messenger/genetics , Rhinitis/genetics , Sinusitis/genetics , Adult , Aged , Cells, Cultured , Eicosanoids , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Lipoxins/biosynthesis , Male , Middle Aged , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasal Polyps/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhinitis/metabolism , Rhinitis/pathology , Sinusitis/metabolism , Sinusitis/pathology
9.
Biochem Pharmacol ; 85(12): 1795-802, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23643932

ABSTRACT

Lipoxin A4 (LXA4) has been described as an anti-inflammatory mediator, which exerts its effects through the formyl peptide receptor FPR2, also known as ALX. However, there has been a controversy whether or not cells expressing FPR2/ALX, such as neutrophils, respond to LXA4. We, therefore, systematically examined the ability of the human and murine forms of the receptor to respond to LXA4. We show that both receptor orthologues responded to the FPR2/ALX peptide agonist WKYMVM when expressed heterologously. In contrast, LXA4 from different sources neither increased [Ca²âº](i) and extracellular-signal-regulated kinase (ERK) phosphorylation, nor did it induce a decrease in cAMP levels or a translocation of ß-arrestin. Also, several LXA4 analogs were found to be unable to signal through FPR2/ALX. We conclude that FPR2/ALX is not activated by LXA4 and that the molecular mechanism by which LXA4 functions still needs to be identified.


Subject(s)
Gene Expression Regulation , Lipoxins/physiology , Receptors, Formyl Peptide/administration & dosage , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Adaptor Proteins, Signal Transducing , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , HEK293 Cells , Humans , Lipoxins/metabolism , Mice , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/biosynthesis
10.
Am J Dermatopathol ; 35(2): 184-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23147350

ABSTRACT

Melanoma, due to its metastatic rate, is among the most aggressive forms of skin cancer. Human formyl peptide receptor (FPR) and its variant FPR-like 1 (FPRL1) have been associated with cell migration and invasiveness in neoplasms. We have studied the in situ expression of these receptors in a large series of melanocytic lesions and correlated the expression with clinicopathological features and prognosis. Tissue microarray blocks of 141 cases including nevi (31 cases), primary (84 cases), and metastatic melanomas (26 cases) were semiquantitatively evaluated by immunohistochemistry for the expression of FPR and FPRL1 proteins. A significant association was observed regarding diagnosis and percentage of cells showing expression of FPR (P = 0.0311) and FPRL1 (P = 0.0053). A gain of FPR immunoreactivity was observed in the lesions having ulceration (P = 0.0194) and Breslow thickness (P = 0.044). Also, high FPRL1 cytoplasmic immunoreactivity was seen in lesions without tumor regression (P = 0.04). In addition, in patients with increased cytoplasmic staining for FPR, the probability of disease-specific survival was significantly lower (log rank test, P = 0.0089). Our findings reveal that FPR and FPRL1 are overexpressed in primary melanoma and correlate with aggressive tumor characteristics, underscoring them as potential therapeutic targets.


Subject(s)
Melanoma/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Skin Neoplasms/metabolism , Biomarkers, Tumor/analysis , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Phenotype , Receptors, Formyl Peptide/analysis , Receptors, Lipoxin/analysis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Tissue Array Analysis
11.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G487-97, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21659618

ABSTRACT

The macrophage plays a major role in the induction and resolution phases of inflammation; however, how lipid mediator-derived signals may modulate macrophage function in the resolution of inflammation driven by microbes (e.g., in inflammatory bowel disease) is not well understood. We examined the effects of aspirin-triggered lipoxin (ATL), a stable analog of lipoxin A(4), on the antimicrobial responses of human peripheral blood mononuclear cell-derived macrophages and the monocytic THP-1 cell line. Additionally, we assessed the expression and localization of the lipoxin receptor, formyl peptide receptor 2 (FPR2), in colonic mucosal biopsies from patients with Crohn's disease to determine whether the capacity for lipoxin signaling is altered in inflammatory bowel disease. We found that THP-1 cells treated with ATL (100 nM) displayed increased phagocytosis of inert fluorescent beads and Escherichia coli in a scavenger receptor- and PI3K-dependent, opsonization-independent manner. This ATL-induced increase in phagocytosis was also observed in primary human macrophages, where it was associated with an inhibition of E. coli-induced IL-1ß and IL-8 production. Finally, we found that FPR2 gene expression was increased approximately sixfold in the colon of patients with Crohn's disease, a finding reproduced in vitro by the treatment of THP-1 cells with interferon-γ or lipopolysaccharide. These results suggest that lipoxin signaling is upregulated in inflammatory environments, and, in addition to their known role in tissue resolution following injury, lipoxins can enhance macrophage clearance of invading microbes.


Subject(s)
Lipoxins/pharmacology , Macrophages/immunology , Aspirin/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/physiology , Crohn Disease/genetics , Crohn Disease/physiopathology , Humans , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Receptors, CCR7/biosynthesis , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Up-Regulation
12.
Int Immunopharmacol ; 11(1): 55-66, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974309

ABSTRACT

Recent studies have associated the dysregulated expression of Annexin-A1/Formyl peptide receptor 2 (FPR2/ALX) system with the development of autoimmune diseases. In this study we systematically scanned human leukocyte subsets for the presence of this pathway aiming to provide a roadmap that will help investigators to explore possible links between the development of immune related disorders and the expression of this system. Our results show that neutrophils, monocytes and NK cells express higher levels of both AnxA1 and FPR2/ALX compared to T or B cells. Further analysis of specific T cell subsets revealed higher levels in activated CD25(+) and memory CD45RO CD4 T cells compared to resting CD25(-) or naïve CD45RA CD4 T cells. Together the results expand our knowledge of the AnxA1-FPR2/ALX system in immune cells and provide new avenues for investigation into the functions of this signalling pathway in systems other than that classically described for neutrophils.


Subject(s)
Annexin A1/biosynthesis , Leukocytes/immunology , Leukocytes/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Adult , Autoimmune Diseases/metabolism , Blotting, Western , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Neutrophils/immunology , Neutrophils/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
13.
J Neurochem ; 113(3): 749-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20141570

ABSTRACT

Recent studies suggest that the chemotactic G protein-coupled receptor formyl-peptide-receptor-like-1 (FPRL1) or the scavenger receptor MARCO (macrophage receptor with collagenous structure) plays an essential role in the inflammatory response of host defense mechanisms and neurodegenerative disorders such as Alzheimer's disease. We therefore analyzed the involvement of FPRL1 and MARCO in amyloid beta1-42 (Abeta1-42)-induced signalling by extracellular-signal regulated kinases 1/2 (ERK1/2) phosphorylation and cAMP level measurement in glial cells (astrocytes and microglia) and in transfected HEK293 cells. Receptors were inhibited by small interference RNA and the consequences in Abeta1-42- and MARCO agonist fucoidan-induced signal transduction were determined. Receptor deactivation by antagonists or small interference RNA verified the importance of FPRL1 for Abeta1-42-mediated signal transduction by ERK1/2 phosphorylation and cAMP level measurement in glial cells. Furthermore, for the first time, we have demonstrated a functional interaction between FPRL1 and scavenger receptors in fucoidan-mediated signalling by ERK1/2 phosphorylation and cAMP level measurement. In addition, co-immunoprecipitation data and fluorescence microscopy measurements revealed a physical interaction between FPR, FPRL1 and MARCO. These results suggest that FPRL1 plays a pivotal role for Abeta1-42-induced signal transduction in glial cells and the interaction with MARCO could explain the broad ligand spectrum of formyl peptide receptors.


Subject(s)
Amyloid beta-Peptides/pharmacology , Neuroglia/drug effects , Peptide Fragments/pharmacology , Receptors, Formyl Peptide/physiology , Receptors, Immunologic/physiology , Signal Transduction/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoprecipitation , Microscopy, Fluorescence , Phosphorylation , Plasmids/genetics , Polysaccharides/pharmacology , RNA/biosynthesis , RNA/isolation & purification , RNA, Small Interfering/genetics , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/drug effects , Receptors, Formyl Peptide/genetics , Receptors, Immunologic/drug effects , Receptors, Immunologic/genetics , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Metabolism ; 59(3): 433-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19850308

ABSTRACT

The systemic inflammatory activity in patients with stable coronary artery disease (CAD) is associated with a dysregulated cortisol response. Moreover, an aberrant activation status of neutrophils in CAD has been discussed; and the question of glucocorticoid resistance has been raised. The anti-inflammatory actions of glucocorticoids are mediated by annexin-1 (ANXA1). We investigated the expression of glucocorticoid receptors (GR) and ANXA1, as well as the exogenous effects of ANXA1 on neutrophils in CAD patients and related the data to diurnal salivary cortisol. Salivary cortisol levels were measured in the morning and evening during 3 consecutive days in 30 CAD patients and 30 healthy individuals. The neutrophil expression of GR and ANXA1 was determined by flow cytometry. The effect of exogenous ANXA1 was determined in a neutrophil stimulation assay. The patients showed a flattened diurnal cortisol pattern compared with healthy subjects, involving higher levels in the evening. The neutrophil expression of GR-total and GR-alpha was decreased, whereas the GR-beta expression did not differ compared with controls. The neutrophil expression of ANXA1 was significantly increased in patients. Ex vivo, ANXA1 impaired the leukotriene B(4)-induced neutrophil production of reactive oxygen species in patients but not in controls. Our findings indicate a persistent overactivation of the hypothalamic-pituitary-adrenal axis in CAD patients but do not give any evidence for glucocorticoid resistance, as assessed by the neutrophil expression of GR and ANXA1. The altered neutrophil phenotype in CAD may thus represent a long-term response to disease-related activation.


Subject(s)
Annexin A1/biosynthesis , Coronary Artery Disease/metabolism , Neutrophils/metabolism , Aged , Angiography , CD18 Antigens/biosynthesis , Circadian Rhythm/physiology , Female , Flow Cytometry , Glucocorticoids/biosynthesis , Glucocorticoids/physiology , Humans , Hydrocortisone/metabolism , Leukotriene B4/pharmacology , Male , Middle Aged , Phenotype , Reactive Oxygen Species , Receptors, Formyl Peptide/biosynthesis , Receptors, Glucocorticoid/biosynthesis , Receptors, Lipoxin/biosynthesis , Saliva/metabolism
15.
J Immunol ; 183(9): 5511-7, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843937

ABSTRACT

We determined the expression of the formyl peptide receptor (FPR) family and the functional roles of the FPR family in NK cells. All tested human NK cells express two members of the FPR family (FPR1 and FPR2). The expression of FPR3 was noted to occur in a donor-specific manner. The stimulation of NK cells with FPR family-selective agonists (fMLF (N-formyl-Met-Leu-Phe), MMK-1, F2L, and WKYMVm (Trp-Lys-Tyr-Met-Val-d-Met)) elicited cytolytic activity in resting NK cells, but not in IL-2-activated NK cells; the cytolytic activity was not inhibited by pertussis toxin. The FPR family agonists also stimulated chemotactic migration of IL-2-activated NK cells, but not resting NK cells; the chemotactic migration was completely inhibited by pertussis toxin. WKYMVm stimulates ERK, p38 MAPK, and JNK activities in both resting and IL-2-activated NK cells. WKYMVm-induced chemotactic migration was partially inhibited by PD98059 (2'-amino-3'-methoxyflavone); however, the inhibition of JNK by its selective inhibitor (SP600125, anthra[1,9-cd]pyrazol-6(2H)-one) dramatically inhibited the WKYMVm-induced cytolytic activity. Furthermore, WKYMVm-induced chemotactic migration and cytolytic activity were partly inhibited by FPR family-selective antagonists (cyclosporin H and WRWWWW). Taken together, our findings indicate that human NK cells express functional members of the FPR family, and in turn the activation of the three members of the FPR receptor family elicit cytolytic activity in NK cells, thus suggesting that the receptors are potentially important therapeutic targets for the modulation of NK cell-mediated immune responses.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Multigene Family/immunology , Receptors, Formyl Peptide/biosynthesis , Chemotaxis, Leukocyte/immunology , Cytotoxicity, Immunologic , Humans , Lymphocyte Activation/immunology , Oligopeptides/agonists , Oligopeptides/biosynthesis , Oligopeptides/physiology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/agonists , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/physiology , Signal Transduction/immunology
16.
J Immunol ; 183(3): 2089-96, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19597002

ABSTRACT

Aspirin is a unique nonsteroidal anti-inflammatory drug; at high doses (aspirin(high), 1g), it is anti-inflammatory stemming from the inhibition of cyclooxygenase and proinflammatory signaling pathways including NF-kappaB, but is cardioprotective at lower doses (aspirin(low), 75 mg). The latter arises from the inhibition of thromboxane (Tx) B(2), a prothrombotic eicosanoid also implicated in polymorphonuclear leukocyte trafficking. As a result, aspirin(low) is widely used as a primary and secondary preventative against vascular disease. Despite this and its ability to synthesize proresolution 15-epi-lipoxin A(4) it is not known whether aspirin(low) is anti-inflammatory in humans. To address this, we generated skin blisters by topically applying cantharidin on the forearm of healthy male volunteers, causing an acute inflammatory response including dermal edema formation and leukocyte trafficking. Although not affecting blister fluid volume, aspirin(low) (75 mg, oral, once daily/10 days) reduced polymorphonuclear leukocyte and macrophage accumulation independent of NF-kappaB-regulated gene expression and inhibition of conventional prostanoids. However, aspirin(low) triggered 15-epi-lipoxin A(4) synthesis and up-regulated its receptor (FPRL1, ALX). From complimentary in vitro experiments, we propose that 15-epi-lipoxin A(4) exerts its protective effects by triggering antiadhesive NO, thereby dampening leukocyte/endothelial cell interaction and subsequent extravascular leukocyte migration. Since similar findings were obtained from murine zymosan-induced peritonitis, we suggest that aspirin(low) possesses the ability to inhibit mammalian innate immune-mediated responses. This highlights 15-epi-lipoxin A(4) as a novel anti-inflammatory working through a defined receptor and suggests that mimicking its mode of action represents a new approach to treating inflammation-driven diseases.


Subject(s)
Aspirin/administration & dosage , Inflammation/drug therapy , Lipoxins/biosynthesis , Acute Disease , Aspirin/pharmacology , Blister/chemically induced , Cantharidin , Cell Adhesion/drug effects , Chemotaxis, Leukocyte/drug effects , Dose-Response Relationship, Drug , Humans , Macrophages , Male , Nitric Oxide/biosynthesis , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Up-Regulation/drug effects
17.
Mol Cancer Res ; 7(6): 907-15, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19491199

ABSTRACT

Emerging evidence suggests that the antimicrobial peptide, leucine leucine-37 (LL-37), could play a role in the progression of solid tumors. LL-37 is expressed as the COOH terminus of human cationic antimicrobial protein-18 (hCAP-18) in ovarian, breast, and lung cancers. Previous studies have shown that the addition of LL-37 to various cancer cell lines in vitro stimulates proliferation, migration, and invasion. Similarly, overexpression of hCAP-18/LL-37 in vivo accelerates tumor growth. However, the receptor or receptors through which these processes are mediated have not been thoroughly examined. In the present study, expression of formyl peptide receptor-like 1 (FPRL1) was confirmed on ovarian cancer cells. Proliferation assays indicated that LL-37 does not signal through a G protein-coupled receptor, such as FPRL1, to promote cancer cell growth. By contrast, FPRL1 was required for LL-37-induced invasion through Matrigel. The peptide stimulated mitogen-activated protein kinase and Janus-activated kinase/signal transducers and activators of transcription signaling cascades and led to the significant activation of several transcription factors, through both FPRL1-dependent and FPRL1-independent pathways. Likewise, expression of some LL-37-stimulated genes was attenuated by the inhibition of FPRL1. Increased expression of CXCL10, EGF, and PDGF-BB as well as other soluble factors was confirmed from conditioned medium of LL-37-treated cells. Taken together, these data suggest that LL-37 potentiates a more aggressive behavior from ovarian cancer cells through its interaction with FPRL1.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Signaling System/physiology , Oncogenes , Ovarian Neoplasms/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Humans , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Lipoxin/antagonists & inhibitors , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/genetics , Recombinant Proteins/pharmacology , Cathelicidins
18.
Mol Pharmacol ; 74(2): 392-402, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18458054

ABSTRACT

Development of immunomodulatory agents that enhance innate immune responses represents a promising strategy for combating infectious diseases. In the present studies, we screened a series of 71 arylcarboxylic acid hydrazide derivatives for their ability to induce macrophage tumor necrosis factor alpha (TNF-alpha) production and identified six such compounds, including one compound previously shown to be a formyl peptide receptor (FPR/FPRL1) agonist. The two most potent compounds [compound 1, nicotinic acid [5-(3-bromophenyl)-2-furyl]methylene-hydrazide; compound 2, 4-fluoro-benzoic acid [5-(3-trifluoromethyl-phenyl)-2-furyl]-methylene-hydrazide] were selected for further analysis. These compounds induced de novo production of TNF-alpha in a dose- and time-dependent manner in human and murine monocyte/macrophage cell lines and in primary macrophages. These compounds also induced mobilization of intracellular Ca(2+), production of reactive oxygen species, and chemotaxis in human and murine phagocytes. Induction of macrophage TNF-alpha production was pertussis toxin-sensitive, and analysis of the cellular target of these compounds showed that they were FPRL1-specific agonists and that this response was blocked by FPR/FPRL1 and FPRL1-specific antagonists. In addition, pharmacophore modeling showed a high degree of similarity for low-energy conformations of these two compounds to the current pharmacophore model for FPR ligands ( Mol Pharmacol 68: 1301-1310, 2005 ). Overall, these compounds represent novel FPRL1 agonists that induce TNF-alpha, a response distinct from those induced by other known FPR and FPRL1 agonists.


Subject(s)
Macrophages/metabolism , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Rats , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis
19.
J Immunol ; 179(8): 5493-503, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911636

ABSTRACT

The proteinase-activated receptors (PARs) are a novel family of G protein-coupled receptors, and their effects in neurodegenerative diseases remain uncertain. Alzheimer's disease (AD) is a neurodegenerative disorder defined by misfolded protein accumulation with concurrent neuroinflammation and neuronal death. We report suppression of proteinase-activated receptor-2 (PAR2) expression in neurons of brains from AD patients, whereas PAR2 expression was increased in proximate glial cells, together with up-regulation of proinflammatory cytokines and chemokines and reduced IL-4 expression (p < 0.05). Glial PAR2 activation increased expression of formyl peptide receptor-2 (p < 0.01), a cognate receptor for a fibrillar 42-aa form of beta-amyloid (Abeta(1-42)), enhanced microglia-mediated proinflammatory responses, and suppressed astrocytic IL-4 expression, resulting in neuronal death (p < 0.05). Conversely, neuronal PAR2 activation protected human neurons against the toxic effects of Abeta(1-42) (p < 0.05), a key component of AD neuropathogenesis. Amyloid precursor protein-transgenic mice, displayed glial fibrillary acidic protein and IL-4 induction (p < 0.05) in the absence of proinflammatory gene up-regulation and neuronal injury, whereas PAR2 was up-regulated at this early stage of disease progression. PAR2-deficient mice, after hippocampal Abeta(1-42) implantation, exhibited enhanced IL-4 induction and less neuroinflammation (p < 0.05), together with improved neurobehavioral outcomes (p < 0.05). Thus, PAR2 exerted protective properties in neurons, but its activation in glia was pathogenic with secretion of neurotoxic factors and suppression of astrocytic anti-inflammatory mechanisms contributing to Abeta(1-42)-mediated neurodegeneration.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/prevention & control , Receptor, PAR-2/physiology , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Astrocytes/metabolism , Cell Line , Cells, Cultured , Female , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Peptide Fragments/toxicity , Rats , Receptor, PAR-2/biosynthesis , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/biosynthesis , Receptors, Lipoxin/genetics
20.
Biomed Pharmacother ; 61(5): 261-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17418999

ABSTRACT

In this study, we investigated the synthesis of lipoxins (LXs) and their anti-inflammatory effects in different human airway epithelial cell culture models. After cell incubation with exogenous 5(S),6(R)-dihydroxy-7,9-trans-11,14-cis-eicosatetraenoic acid, LXA(4) was detected in supernatants of differentiated human bronchial epithelial cells by contrast to non-differentiated cells. Exogenous LXA(4) significantly inhibited tumor necrosis factor-alpha (TNF-alpha)-induced interleukin-8 (IL-8) release in the different epithelial cell types and the potency of inhibition was dependent of the accessibility of the specific LXA(4) receptor, formyl-peptide receptor like-1 (FPRL-1) expressed by all these cells. Immunohistochemistry analysis on human bronchial biopsies showed a high expression of FPRL-1 in the epithelium. Finally, an FPRL-1 receptor antagonist, boc-2 peptide reversed LXA(4) effect on IL-8 generation. Together, these findings indicate that differentiated human bronchial epithelium synthesizes LX in vivo which could have autocrine actions through its specific receptor FPRL-1 to promote resolution of airway inflammation.


Subject(s)
Epithelial Cells/metabolism , Lipoxins/biosynthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Bronchi/cytology , Cells, Cultured , Chromatography, High Pressure Liquid , Epithelial Cells/drug effects , Humans , Hydroxyeicosatetraenoic Acids/pharmacology , Immunochemistry , Interleukin-8/biosynthesis , Lipoxins/pharmacology , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Respiratory Mucosa/cytology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...