Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
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.
Biochim Biophys Acta ; 1861(9 Pt A): 1025-1035, 2016 09.
Article in English | MEDLINE | ID: mdl-27317426

ABSTRACT

Resolution of inflammation is an active process involving a novel category of lipid factors known as specialized pro-resolving lipid mediators, which includes Resolvin D1 (RvD1). While accumulating evidence suggests that RvD1 counteracts proinflammatory signaling and promotes resolution, the specific cellular targets and mechanisms of action of RvD1 remain largely unknown. In the present study, we investigated the role and molecular mechanisms of RvD1 in ischemia/reperfusion (IR)-induced sterile liver inflammation. Male C57BL/6 mice underwent 70% hepatic ischemia for 60min, followed by reperfusion. RvD1 (5, 10, and 15µg/kg, i.p.) was administered to the mice 1h before ischemia and then immediately prior to reperfusion. RvD1 attenuated IR-induced hepatocellular damage and the proinflammatory response. In purified Kupffer cells (KCs) from mice exposed to IR, the levels of M1 marker genes (Nos2a and Cd40) increased, while those of M2 marker genes (Arg1, Cd206, and Mst1r) decreased, demonstrating a proinflammatory shift. RvD1 markedly attenuated these changes. Depletion of KCs by liposome clodronate abrogated the effects of RvD1 on proinflammatory mediators and macrophage polarization. In addition, RvD1 attenuated increases in myeloperoxidase activity and Cxcl1 and Cxcl2 mRNA expression. RvD1 markedly augmented the efferocytic activity of KCs, as indicated by increases in F4/80(+)Gr-1(+) cells in the liver. However, antagonist pretreatment or gene silencing of the RvD1 receptor, ALX/FPR2, abrogated the anti-inflammatory and pro-resolving actions of RvD1. These data indicate that RvD1 ameliorates IR-induced liver injury, and this protection is associated with enhancement of M2 polarization and efferocytosis via ALX/FPR2 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Docosahexaenoic Acids/administration & dosage , Inflammation/drug therapy , Receptors, Formyl Peptide/genetics , Reperfusion Injury/drug therapy , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Arginase/genetics , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/metabolism , Inflammation/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Lectins, C-Type/genetics , Liver/injuries , Liver/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Phagocytosis/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/genetics , Receptors, Formyl Peptide/biosynthesis , Reperfusion Injury/pathology , Signal Transduction
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 ; 195(3): 1139-51, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101324

ABSTRACT

Blood-derived monocytes remove apoptotic cells and terminate inflammation in settings as diverse as atherosclerosis and Alzheimer's disease. They express high levels of the proresolving receptor ALX/FPR2, which is activated by the protein annexin A1 (ANXA1), found in high abundance in inflammatory exudates. Using primary human blood monocytes from healthy donors, we identified ANXA1 as a potent CD14(+)CD16(-) monocyte chemoattractant, acting via ALX/FPR2. Downstream signaling pathway analysis revealed the p38 MAPK-mediated activation of a calcium independent phospholipase A2 with resultant synthesis of lysophosphatidic acid (LPA) driving chemotaxis through LPA receptor 2 and actin cytoskeletal mobilization. In vivo experiments confirmed ANXA1 as an independent phospholipase A2-dependent monocyte recruiter; congruently, monocyte recruitment was significantly impaired during ongoing zymosan-induced inflammation in AnxA1(-/-) or alx/fpr2/3(-/-) mice. Using a dorsal air-pouch model, passive transfer of apoptotic neutrophils between AnxA1(-/-) and wild-type mice identified effete neutrophils as the primary source of soluble ANXA1 in inflammatory resolution. Together, these data elucidate a novel proresolving network centered on ANXA1 and LPA generation and identify previously unappreciated determinants of ANXA1 and ALX/FPR2 signaling in monocytes.


Subject(s)
Annexin A1/immunology , Apoptosis/immunology , Monocytes/immunology , Neutrophils/immunology , Receptors, Lysophosphatidic Acid/immunology , Actin Cytoskeleton/metabolism , Animals , Annexin A1/genetics , Cells, Cultured , Enzyme Activation/immunology , Humans , Inflammation/immunology , Lipopolysaccharide Receptors/metabolism , Lysophospholipids/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/transplantation , Phospholipases A2, Calcium-Independent/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, IgG/metabolism , Receptors, Lysophosphatidic Acid/genetics , Zymosan , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Anticancer Res ; 35(5): 2769-74, 2015 May.
Article in English | MEDLINE | ID: mdl-25964556

ABSTRACT

BACKGROUND/AIM: Formyl-peptide receptors (FPRs) are expressed in several tissues and cell types. The identification of markers involved in cell growth may further allow for molecular profiling of lung cancer. We investigated the possible role of FPRs as molecular markers in several types of lung carcinomas which is the main cause of cancer death worldwide. MATERIALS AND METHODS: Tumor tissue samples were collected from six patients affected by lung cancer. Biopsies were analyzed for expression of FPR isoforms both in tumoral and peritumoral tissue by real-time polymerase chain reaction (PCR), western blot and immunofluorescence. RESULTS: Real-time PCR, western blot and immunofluorescence analyses showed that FPR expression is lower in types of human lung cancer tissues when compared to the surrounding peritumoral tissues. CONCLUSION: The study of the mechanistic basis for the control of FPR expression in normal peritumoral versus tumoral tissues could provide the basis for new diagnostic and therapeutic interventions.


Subject(s)
Biomarkers, Tumor/biosynthesis , Lung Neoplasms/genetics , Receptors, Formyl Peptide/biosynthesis , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Protein Isoforms/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Formyl Peptide/genetics
8.
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
9.
Anticancer Res ; 34(5): 2223-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24778024

ABSTRACT

BACKGROUND: Formyl peptide receptor 1 (FPR1) as a regulator of innate inflammatory response has been implicated in tumor progression of gliomas. The purpose of the present study was to evaluate the prognostic significance and the ligand-receptor interaction of FPR1 in gastric cancer (GC). PATIENTS AND METHODS: FPR1 was immunohistochemically-analyzed in tissue sections originating from 116 GC patients. Reverse transcription-polymerase chain reaction (RT-PCR) was used for the assessment of interaction between FPR1 and the FPR1 ligand annexin A1 (AnxA1) in GC cells. RESULTS: High FPR1 expression was significantly associated with stage IV disease, submucosal invasion, serosal invasion, and clinical outcome of GC. Multivariate analysis showed that high FPR1 expression was an independent risk factor of poor overall survival in GC patients. FPR1 expression increased significantly when AnxA1 overexpression was present in GC cells. A positive feedback regulation of FPR1 was involved in the AnxA1-FPR1 signal transduction. CONCLUSION: FPR1 expression may be used as a novel indicator to predict outcome in GC patients after gastrectomy.


Subject(s)
Biomarkers, Tumor/analysis , Receptors, Formyl Peptide/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Gastrectomy/mortality , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
10.
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
11.
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
12.
Protein Pept Lett ; 20(11): 1272-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23746112

ABSTRACT

Human N-formyl peptide receptors (FPRs) belong to the G protein-coupled receptors (GPCRs) superfamily, the most frequently addressed drug targets in the pharmaceutical industry, and are considered to play important roles in innate immunity and host defense mechanisms. Although still a highly challenging task, the availability of soluble and functional GPCRs including FPRs in milligram quantities is essential to spur the advancement of protein-based structural and functional studies for drug discovery. In this report, the applicability of E. coli extracts-based cell-free expression system to producing soluble and active human FPRs and hence to FPRs protein-based research was evaluated, during which human FPR3 was selected as our prototype receptor. To better solubilize the freshly expressed human FPR3, a panel of different detergents (mostly nonionic detergents) were selected and evaluated in the cell-free system devoid of natural membrane. After one-step immunoaffinity purification, the secondary structure and biological function of purified FPR3 were characterized. A final yield of 0.6 mg functional human FPR3 per ml reaction volume was obtained. The demonstrated proper folding and functionality of the cell-free produced human FPR3 opens a new avenue for the fast and efficient generation of human FPRs (and even other GPCRs) for structural and functional analysis.


Subject(s)
Cell-Free System , Escherichia coli/genetics , Receptors, Formyl Peptide/genetics , Receptors, G-Protein-Coupled/biosynthesis , Humans , Protein Structure, Secondary , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/chemistry , Receptors, G-Protein-Coupled/genetics , Structure-Activity Relationship
13.
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
14.
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
15.
J Immunol ; 188(4): 1799-808, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22262660

ABSTRACT

Formyl peptide receptors (FPRs) are chemoattractant receptors that mediate inflammatory cell responses to infection. Recent evidence indicates that noneosinophilic asthma phenotypes can be developed by both Th1 and Th17 cell responses when exposed to LPS-containing allergens. In this study, we evaluated the effects of airway activation of FPRs by their synthetic agonist, Trp-Lys-Tyr-Met-Val-D-Met (W-peptide), on the development of Th1 and Th17 cell responses in a noneosinophilic asthma mouse model. A noneosinophilic asthma mouse model was generated by intranasal sensitization with 10 µg of LPS plus 75 µg of OVA on days 0, 1, 2, and 7. Mice were then challenged with 50 µg of OVA alone on days 14, 15, 21, and 22. W-peptide was administered during the sensitization period, and immune and inflammatory responses were evaluated after OVA challenge. Lung inflammation after OVA challenge was partly abolished by airway activation of FPRs during sensitization. Maturation of dendritic cells (DCs) and migration of DCs from the lung to lung-draining lymph nodes were inhibited by FPR activation. In addition, airway activation of FPRs inhibited allergen-specific T cell proliferation in the lymph nodes. Production of IL-12 and IL-6 (Th1- and Th17-polarizing cytokines) from lung DCs was decreased by airway activation of FPRs. This effect resulted in the inhibition of allergen-specific Th1 and Th17 cell responses. Airway activation of FPRs during sensitization effectively prevents the development of Th1 and Th17 cell responses induced by LPS-containing allergens via multiple mechanisms, such as inhibition of DC maturation and migration and the production of Th1- and Th7-polarizing cytokines.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Lung/immunology , Receptors, Formyl Peptide/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Asthma/metabolism , Asthma/pathology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Disease Models, Animal , Interleukin-12/biosynthesis , Interleukin-6/biosynthesis , Lung/metabolism , Lung/pathology , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Ovalbumin/immunology , Receptors, Formyl Peptide/biosynthesis , Th1 Cells/drug effects , Th17 Cells/drug effects
16.
Neurosci Lett ; 506(2): 266-70, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22133809

ABSTRACT

Pattern recognition receptors (PRRs) play important roles in the inflammatory responses to Alzheimer's disease (AD). Our previous study indicated that soybean isoflavone (SIF) exhibited anti-inflammatory effect in rats treated by ß-amyloid peptides1-42 (Aß1-42). In present study, we further detected the effects of SIF against inflammation caused by Aß1-42 treatment in rats. Serum inflammatory mediators and neurotrophic factors including transforming growth factor-ß (TGF-ß), inducible nitric oxide synthase (iNOS), brain-derived neurotrophic factor (BDNF) and S100ß were detected by enzyme-like immunosorbent assay (ELISA). Reverse transcription-polymerase chain reaction (RT-PCR) and western blot methods were applied for detecting mRNA and protein expression of interleukin-1ß (IL-1ß), iNOS, tumor necrosis factor-α (TNF-α), TGF-ß, BDNF, S100ß, myeloid differentiation factor88 (Myd88), Toll-like receptor2 (TLR2), formyl peptide receptors (FPRs), inhibitor κB kinase (IKK) and inhibitor κB-α (IκB-α) in rat's brain tissue. Our results indicated that SIF could reduce the production of IL-1ß, TNF-α and iNOS induced by Aß1-42 in serum and brain of rats. SIF also significantly reversed Aß1-42-induced up-regulation of TLR2, FPR, Myd88, IKK and decreased IκB-α mRNA and protein expressions in rats. These results suggested that TLR2 and FPR might involve in the inflammatory process induced by Aß1-42 treatment, and SIF was an efficiency compound in reversing the inflammation caused by Aß1-42 treatment.


Subject(s)
Amyloid beta-Peptides/toxicity , Glycine max/chemistry , Isoflavones/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Receptors, Pattern Recognition/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Inflammation/chemically induced , Inflammation/metabolism , Male , Rats , Rats, Wistar , Receptors, Formyl Peptide/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2/biosynthesis
17.
PLoS One ; 6(12): e28712, 2011.
Article in English | MEDLINE | ID: mdl-22174875

ABSTRACT

The gene encoding the human formyl peptide receptor 1 (FPR1) is heterogeneous, containing numerous single nucleotide polymorphisms (SNPs). Here, we examine the effect of these SNPs on gene transcription and protein translation. We also identify gene promoter sequences and putative FPR1 transcription factors. To test the effect of codon bias and codon pair bias on FPR1 expression, four FPR1 genetic variants were expressed in human myeloid U937 cells fused to a reporter gene encoding firefly luciferase. No significant differences in luciferase activity were detected, suggesting that the translational regulation and protein stability of FPR1 are modulated by factors other than the SNP codon bias and the variant amino acid properties. Deletion and mutagenesis analysis of the FPR1 promoter showed that a CCAAT box is not required for gene transcription. A -88/41 promoter construct resulted in the strongest transcriptional activity, whereas a -72/41 construct showed large reduction in activity. The region between -88 and -72 contains a consensus binding site for the transcription factor PU.1. Mutagenesis of this site caused significant reduction in reporter gene expression. The PU.1 binding was confirmed in vivo by chromatin immunoprecipitation, and the binding to nucleotides -84 to -76 (TTCCTATTT) was confirmed in vitro by an electrophoretic mobility shift assay. Thus, similar to many other myeloid genes, FPR1 promoter activity requires PU.1. Two single nucleotide polymorphisms at -56 and -54 did not significantly affect FPR1 gene expression, despite differences in binding of transcription factor IRF1 in vitro. Inflammatory mediators such as interferon-γ, tumor necrosis factor-α, and lipopolysaccharide did not increase FPR1 promoter activity in myeloid cells, whereas differentiation induced by DMSO and retinoic acid enhanced the activity. This implies that the expression of FPR1 in myeloid cells is developmentally regulated, and that the differentiated cells are equipped for immediate response to microbial infections.


Subject(s)
Inflammation Mediators/metabolism , Polymorphism, Single Nucleotide/genetics , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Transcription Factors/metabolism , Base Sequence , Binding Sites , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Codon/genetics , Dimethyl Sulfoxide/pharmacology , Gene Expression Regulation/drug effects , Genotype , Humans , Molecular Sequence Data , Mutagenesis/drug effects , Mutagenesis/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Reproducibility of Results , Transcription, Genetic/drug effects , Tretinoin/pharmacology , U937 Cells
18.
PLoS One ; 6(8): e23076, 2011.
Article in English | MEDLINE | ID: mdl-21853070

ABSTRACT

G-protein coupled receptors (GPCRs) participate in a wide range of vital regulations of our physiological actions. They are also of pharmaceutical importance and have become many therapeutic targets for a number of disorders and diseases. Purified GPCR-based approaches including structural study and novel biophysical and biochemical function analyses are increasingly being used in GPCR-directed drug discovery. Before these approaches become routine, however, several hurdles need to be overcome; they include overexpression, solubilization, and purification of large quantities of functional and stable receptors on a regular basis. Here we report milligram production of a human formyl peptide receptor 3 (FPR3). FPR3 comprises a functionally distinct GPCR subfamily that is involved in leukocyte chemotaxis and activation. The bioengineered FPR3 was overexpressed in stable tetracycline-inducible mammalian cell lines (HEK293S). After a systematic detergent screening, fos-choline-14 (FC-14) was selected for subsequent solubilization and purification processes. A two-step purification method, immunoaffinity using anti-rho-tag monoclonal antibody 1D4 and gel filtration, was used to purify the receptors to near homogeneity. Immunofluorescence analysis showed that expressed FPR3 was predominantly displayed on cellular membrane. Secondary structural analysis using circular dichroism showed that the purified FPR3 receptor was correctly folded with >50% α-helix, which is similar to other known GPCR secondary structures. Our method can readily produce milligram quantities of human FPR3, which would facilitate in developing human FPR as therapeutic drug targets.


Subject(s)
Bioengineering/methods , Receptors, Formyl Peptide/biosynthesis , Cell Membrane/drug effects , Cell Membrane/metabolism , Chromatography, Affinity , Chromatography, Gel , Circular Dichroism , Detergents/pharmacology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Protein Structure, Secondary , Protein Transport/drug effects , Receptors, Formyl Peptide/isolation & purification , Solubility/drug effects
19.
J Immunol ; 187(3): 1475-85, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709160

ABSTRACT

The peptide F2L was previously characterized as a high-affinity natural agonist for the human formyl peptide receptor (FPR) 3. F2L is an acetylated 21-aa peptide corresponding with the N terminus of the intracellular heme-binding protein 1 (HEBP1). In the current work, we have investigated which proteases were able to generate the F2L peptide from its precursor HEBP1. Structure-function analysis of F2L identified three amino acids, G(3), N(7), and S(8), as the most important for interaction of the peptide with FPR3. We expressed a C-terminally His-tagged form of human HEBP1 in yeast and purified it to homogeneity. The purified protein was used as substrate to identify proteases generating bioactive peptides for FPR3-expressing cells. A conditioned medium from human monocyte-derived macrophages was able to generate bioactivity from HEBP1, and this activity was inhibited by pepstatin A. Cathepsin D was characterized as the protease responsible for HEBP1 processing, and the bioactive product was identified as F2L. We have therefore determined how F2L, the specific agonist of FPR3, is generated from the intracellular protein HEBP1, although it is unknown in which compartment the processing by cathepsin D occurs in vivo.


Subject(s)
Carrier Proteins/metabolism , Cathepsin D/physiology , Chemotactic Factors/agonists , Hemeproteins/metabolism , Peptides/agonists , Protein Precursors/metabolism , Protein Processing, Post-Translational/immunology , Receptors, Formyl Peptide/metabolism , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/biosynthesis , Cathepsin D/deficiency , Cells, Cultured , Chemotactic Factors/biosynthesis , Chemotactic Factors/metabolism , Cricetinae , Cricetulus , Heme-Binding Proteins , Hemeproteins/biosynthesis , Humans , Ligands , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Peptides/metabolism , Protein Binding/immunology , Protein Precursors/biosynthesis , Receptors, Formyl Peptide/biosynthesis
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...