Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Dis Markers ; 2022: 1666240, 2022.
Article in English | MEDLINE | ID: mdl-35692878

ABSTRACT

Exploring the role of neuropeptides in the communication between monocyte subtypes facilitates an investigation of the pathogenesis of Kawasaki disease (KD). We investigated the patterns of interaction between neuropeptide-associated ligands and receptors in monocyte subpopulations in KD patients. Single-cell analysis was employed for the identification of cell subpopulations in KD patients, and monocytes were classified into 3 subpopulations: classical monocytes (CMs), intermediate monocytes (IMs), and nonclassical monocytes (NCMs). Cell-cell communication and differential analyses were used to identify ligand-receptor interactions in monocytes. Five neuropeptide-related genes (SORL1, TNF, SORT1, FPR2, and ANXA1) were involved in cell-cell interactions, wherein FPR2, a neuropeptide receptor, was significantly highly expressed in KD. Weighted gene coexpression network analysis revealed a significant correlation between the yellow module and FPR2 (p < 0.001, CC = 0.43). Using the genes in the yellow module, we constructed a PPI network to assess the possible functions of the FPR2-associated gene network. Gene set enrichment analysis showed that increased FPR2 levels may be involved in immune system regulation. FPR2 in CMs mediates the control of inflammation in KD. The findings of this study may provide a novel target for the clinical treatment of KD.


Subject(s)
Monocytes , Mucocutaneous Lymph Node Syndrome , Computational Biology , Humans , LDL-Receptor Related Proteins , Membrane Transport Proteins , Monocytes/metabolism , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/pathology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Receptors, Neuropeptide , Single-Cell Analysis
2.
Bioengineered ; 13(2): 4028-4038, 2022 02.
Article in English | MEDLINE | ID: mdl-35129068

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a progressive degenerative disease, of which smoking is the main causer. We carried out this study with the aim of exploring the underlying mechanism of methylprednisolone (MP) treating the COPD. To stimulate COPD in vitro, cigarette smoke extract (CSE)was employed to induce human bronchial epithelial cells BEAS-2B. With the help of MTT and Tunel assays, the viability and apoptosis of BEAS-2B cells after indicated treatment were assessed. The levels of inflammatory response and oxidative stress were determined by the changes of markers basing on their commercial kits. Additionally, annexin A1 (ANXA1) expressions at both protein and mRNA levels were assessed with Western blot and Reverse transcription­quantitative PCR (RT-qPCR). Moreover, the expressions of apoptosis- and formyl peptide receptor 2 (FPR2) receptors and the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway-related proteins were determined with Western blot., related proteins and proteins. As a result, MP up-regulated the ANXA1 expression in CSE-induced BEAS-2B cells. MP enhanced the viability but suppressed the apoptosis, inflammatory response and oxidative stress of CSE-induced BEAS-2B cells via regulating FPR2/AMPK pathway, while ANXA1 knockdown exhibited oppositive effects on them. In conclusion, MP up-regulated ANXA1 to inhibit the inflammation, apoptosis and oxidative stress of BEAS-2B cells induced by CSE, alleviating COPD through suppressing the FPR2/AMPK pathway.


Subject(s)
Annexin A1/genetics , Methylprednisolone/pharmacology , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Smoke/adverse effects , AMP-Activated Protein Kinases/genetics , Apoptosis/drug effects , Bronchi/cytology , Cell Line , Epithelial Cells/drug effects , Humans , Inflammation/metabolism , Models, Biological , Oxidative Stress/drug effects , Signal Transduction/drug effects , Nicotiana , Up-Regulation/drug effects
3.
J Invest Dermatol ; 142(8): 2238-2248.e8, 2022 08.
Article in English | MEDLINE | ID: mdl-34979109

ABSTRACT

Chronic wounds represent a major health problem worldwide. Some of the available therapies based on recombinant proteins usually fail owing to the hostile environment found at the wound bed. Aptamers appear as an attractive alternative to recombinant factors owing in part to their stability, sensitivity, specificity, and low-cost production. In this study, the Cell-Systematic Evolution of Ligands by EXponential Enrichment technology was employed to generate aptamers that specifically recognize and modulate the function of the FPR2, a receptor expressed in a variety of cells involved in wound repair. Three aptamers were obtained that specifically bound to FPR2 stable transfectants generated in HaCaT cells. The targeted aptamers were shown to act as FPR2 agonists in different in vitro functional assays, including wound healing assays, and elicited a similar pattern of response to that obtained with other known FPR2 peptide agonists, such as the human LL37 cathelicidin. We have also obtained in vivo evidence for the prohealing activities of one of these FPR2 aptamers in a skin-humanized mouse model developed by us, previously shown to accurately recreate the main phases of physiological human wound repair process. In conclusion, we provide evidence of the potential therapeutic value of FPR2 aptamers for cutaneous repair.


Subject(s)
Aptamers, Nucleotide , Receptors, Formyl Peptide , Animals , Humans , Ligands , Mice , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/agonists , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Wound Healing
4.
J Assist Reprod Genet ; 39(1): 239-250, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35018584

ABSTRACT

PURPOSE: The dysfunction of trophoblast during inflammation plays an important role in PE. Formyl peptide receptor 2 (FPR2) plays crucial roles in the development of inflammation-associated disease. This present study aimed to explore the effect of FPR2 on a trophoblast cellular model of preeclampsia. METHODS: The expression of FPR2 in placenta was detected by immunohistochemical staining and western blotting. Transfection of siRNA was used to knockdown FPR2 in HTR-8/SVneo cells. Inflammatory cytokines were detected by ELISA. CCK8, Transwell, wound healing, FACS and tube formation assays were performed to observe the abilities of cell proliferation, migration, invasion, apoptosis and angiogenesis. Western blotting was implemented to clarify that NF-κB signaling pathway was downstream of FPR2. RESULTS: The expression levels of FPR2 were higher in placental tissues of patients with PE. Knockdown of FPR2 expression by siFPR2 or inhibition of its activity by WRW4 decreased the release of proinflammatory cytokines in HTR8/SVneo cells treated with LPS. Knockdown of FPR2 expression or inhibition of its activity further reversed the LPS-induced attenuation of the proliferation, migration, invasion and angiogenesis and increase in apoptosis in HTR8/SVneo cells. Moreover, the NF-κB signaling pathway was activated in both placental tissues of patients with PE and LPS-treated HTR8/SVneo cells. However, the activation was attenuated when FPR2 was knocked down or inhibited. CONCLUSION: Suppression of FPR2 expression alleviated the effects of inflammation induced by LPS on trophoblasts via the NF-κB signaling pathway, which provided a novel and potential strategy for the treatment of PE.


Subject(s)
Gene Expression/physiology , Inflammation/prevention & control , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Trophoblasts/metabolism , Adult , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Inflammation/physiopathology , NF-kappa B/antagonists & inhibitors , Pre-Eclampsia/drug therapy , Pre-Eclampsia/physiopathology , Pregnancy , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics
5.
J Pharmacol Sci ; 148(1): 56-64, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924130

ABSTRACT

Acute kidney injury (AKI) is a serious complication in critically ill patients. Accumulating evidences indicated that macrophages play an important pro-inflammatory role in AKI and isoliquiritigenin (ISL) can inhibit macrophagic inflammation, but its role in AKI and the underlying mechanism are unknown. The present study aims to investigate the renoprotective effect of ISL on AKI and the role of Formyl peptide receptors 2 (FPR2) in this process. In this study, cisplatin-induced AKI model and lipopolysaccharide-induced macrophage inflammatory model were employed to perform the in vivo and in vitro experiments. The results showed that ISL strongly relieved kidney injury and inhibited renal inflammation in vivo and suppress macrophagic inflammatory response in vitro. Importantly, it was found that FPR2 was significantly upregulated compared to the control group in AKI and LPS-induced macrophage, whereas it was strongly suppressed by ISL. Interestingly, overexpression of FPR2 with transfection of pcDNA3.1-FPR2 effectively reversed the anti-inflammatory effect of ISL in macrophage, suggesting that FPR2 may be the potential target for ISL to prevent inflammation and improve kidney injury of AKI. Take together, these findings indicated that ISL improved cisplantin-induced kidney injury by inhibiting FPR2 involved macrophagic inflammation, which may provide a potential therapeutic option for AKI.


Subject(s)
Acute Kidney Injury/genetics , Acute Kidney Injury/prevention & control , Chalcones/pharmacology , Chalcones/therapeutic use , Cisplatin/adverse effects , Macrophages/metabolism , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Animals , Cells, Cultured , Chalcones/isolation & purification , Gene Expression/drug effects , Glycyrrhiza/chemistry , Inflammation , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Phytotherapy , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Receptors, Lipoxin/physiology , Up-Regulation/drug effects
6.
J Ovarian Res ; 14(1): 177, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34930387

ABSTRACT

BACKGROUND: In our previous study, we found that formyl peptide receptor 2 (FPR2) promoted the invasion and metastasis of epithelial ovarian cancer (EOC) and could be a prognostic marker for EOC. In this study, we aimed to study the possible mechanism of FPR2 in promoting EOC progression. METHODS: EOC cell lines with ectopic FPR2 expression and knockdown as well as their control cell lines were established, and the expression change of RhoA in each cell line was evaluated by real time quantitative polymerase chain reaction (RT-qPCR) and Western blot. Wound healing and Transwell assays were performed to detect the migratory ability of EOCs affected by FPR2 and RhoA. The supernatant of each EOC cell line was used to coculture with macrophages, and then we tested M1 and M2 macrophage biomarkers in the supernatants by flow cytometry. The THP-1 cell line was also induced to differentiate into M1 and M2 macrophages, and FPR2 and RhoA expression in each macrophage cell line was detected by RT-qPCR and Western blot. A tumour xenograft model was established with SKOV3 and SKOV3-shFPR2 cell lines, and tumour volumes and weights were recorded. RESULTS: RhoA expression was significantly increased in EOCs along with the overexpression of FPR2, which showed a positive correlation by Pearson correlation analysis. Ectopic FPR2 expression contributes to the migratory ability of EOCs, and a RhoA inhibitor (C3 transferase) impairs EOC migration. Furthermore, FPR2 stimulated the secretion of Th2 cytokines by EOCs, which induced macrophages to differentiate to the M2 phenotype, while a RhoA inhibitor stimulated the secretion of Th1 cytokines and induced macrophages to differentiate to the M1 phenotype. Moreover, compared with M1 macrophages and THP-1 cells, FPR2 and RhoA expression was significantly upregulated in M2 macrophages. CONCLUSION: FPR2 stimulated M2 macrophage polarization and promoted invasion and metastasis of ovarian cancer cells through RhoA.


Subject(s)
Biomarkers, Tumor , Carcinoma, Ovarian Epithelial , Macrophages/immunology , Ovarian Neoplasms , Receptors, Formyl Peptide , Receptors, Lipoxin , rhoA GTP-Binding Protein , ADP Ribose Transferases/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Botulinum Toxins/pharmacology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line , Cell Movement/drug effects , Cytokines/immunology , Disease Progression , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
7.
Bioorg Chem ; 116: 105338, 2021 11.
Article in English | MEDLINE | ID: mdl-34521045

ABSTRACT

Four pairs of novel meroterpenoid dimers, (±)-applandimeric acids A-D (1-4) with an unprecedented spiro[furo[3,2-b]benzofuran-3,2'-indene] core were isolated from the fruiting bodies of Ganoderma applanatum. Their planar structures were unambiguously determined via extensive spectroscopic analysis. Their relative and absolute configurations were confirmed through calculated internuclear distance, coupling constant, 13C NMR with DP4 + analysis and electronic circular dichroism (ECD). Furthermore, the molecular docking-based method was used to evaluate their interaction with formyl peptide receptor 2 (FPR2) associated with inflammation. Interestingly, (±)-applandimeric acid D (4) can bond with FPR2 by some key hydrogen bonds. Furthermore, an in vitro bioassay verified that 4 can inhibit the expression of FPR2 with IC50 value of 7.93 µM. In addition, compared to the positive control LiCl (20 mM), 4 showed comparable anti-lipogenesis activity at the concentration of 20 µM. Meanwhile, 4 can suppress the protein levels of peroxisome proliferators-activated receptor-γ (PPAR-γ), CCAAT/enhancer-binding protein-ß (C/EBP-ß), adipocyte fatty acid-binding protein 4 (FABP4), and fatty acid synthase (FAS) through activating AMP-activated protein kinase (AMPK) signaling pathway. Thus, our findings indicate that compound 4 could be a lead compound to treat obesity and obesity-related diseases by inhibiting lipid accumulation in adipocyte and alleviating inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ganoderma/chemistry , Lipogenesis/drug effects , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Terpenes/pharmacology , 3T3-L1 Cells , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Mice , Molecular Dynamics Simulation , Molecular Structure , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Structure-Activity Relationship , Terpenes/chemistry , Terpenes/isolation & purification
8.
J Histochem Cytochem ; 69(8): 523-534, 2021 08.
Article in English | MEDLINE | ID: mdl-34339312

ABSTRACT

Radiation therapy-mediated salivary gland destruction is characterized by increased inflammatory cell infiltration and fibrosis, both of which ultimately lead to salivary gland hypofunction. However, current treatments (e.g., artificial saliva and sialagogues) only promote temporary relief of symptoms. As such, developing alternative measures against radiation damage is critical for restoring salivary gland structure and function. One promising option for managing radiation therapy-mediated damage in salivary glands is by activation of specialized proresolving lipid mediator receptors due to their demonstrated role in resolution of inflammation and fibrosis in many tissues. Nonetheless, little is known about the presence and function of these receptors in healthy and/or irradiated salivary glands. Therefore, the goal of this study was to detect whether these specialized proresolving lipid mediator receptors are expressed in healthy salivary glands and, if so, if they are maintained after radiation therapy-mediated damage. Our results indicate that specialized proresolving lipid mediator receptors are heterogeneously expressed in inflammatory as well as in acinar and ductal cells within human submandibular glands and that their expression persists after radiation therapy. These findings suggest that epithelial cells as well as resident immune cells represent potential targets for modulation of resolution of inflammation and fibrosis in irradiated salivary glands.


Subject(s)
Radiation Tolerance , Receptors, Chemokine/genetics , Receptors, Formyl Peptide/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Leukotriene B4/genetics , Receptors, Lipoxin/genetics , Submandibular Gland/radiation effects , Acinar Cells/cytology , Acinar Cells/metabolism , Acinar Cells/radiation effects , Adult , Aged , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Female , Gamma Rays , Gene Expression , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Male , Middle Aged , Receptors, Chemokine/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Leukotriene B4/metabolism , Receptors, Lipoxin/metabolism , Submandibular Gland/cytology , Submandibular Gland/metabolism
9.
Inflamm Res ; 70(3): 309-321, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33502586

ABSTRACT

OBJECTIVE AND DESIGN: This study is aimed at uncovering the signaling pathways activated by vasoactive intestinal peptide in human macrophages MATERIALS: Human peripheral blood mononuclear cell-derived macrophages were used for the in vitro investigation of the VIP-activated signaling pathways. METHODS AND TREATMENT: Time-course and dose-response experiments and siRNA were used in human macrophages co-challenged with various concentrations of VIP and different MAPK pharmacologic inhibitors to investigate signaling pathways activated by VIP. Flow analysis was performed to assess the levels of CD11b, CD35 and CD66. Luminescence spectrometry was used to measure the levels of the released hydrogen peroxide and the intracellular calcium levels in the media. RESULTS: Macrophages incubated with VIP showed increased phospho-AKT and phospho-ERK1/2 levels in a GTP-RhoA-GTPase-dependent manner. Similarly, VIP increased intracellular release of H2O2 and calcium via PLC and GTP-RhoA-GTPase, in addition to inducing the expression of CD11b, CD35, CD66 and MMP9. Furthermore, VIP activated P38 MAPK through the cAMP/PKA pathway but was independent of both PLC and RhoA signaling. The above-mentioned VIP effects were mediated via activation of the FPRL1 receptor. CONCLUSION: VIP/FPRL1/VPAC/GTP-RhoA-GTPase signaling modulated macrophages phenotype through activation of multiple signaling pathways including ERK1/2, AKT, P38, ROS, cAMP and calcium.


Subject(s)
Macrophages/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Vasoactive Intestinal Peptide/metabolism , rhoA GTP-Binding Protein/metabolism , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hydrogen Peroxide/metabolism , Matrix Metalloproteinase 9/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Signal Transduction , Type C Phospholipases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/genetics
10.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118849, 2020 12.
Article in English | MEDLINE | ID: mdl-32916203

ABSTRACT

FPR2, a member of the family of G protein-coupled receptors (GPCRs), mediates neutrophil migration, a response that has been linked to ß-arrestin recruitment. ß-Arrestin regulates GPCR endocytosis and can also elicit non-canonical receptor signaling. To determine the poorly understood role of ß-arrestin in FPR2 endocytosis and in NADPH-oxidase activation in neutrophils, Barbadin was used as a research tool in this study. Barbadin has been shown to bind the clathrin adaptor protein (AP2) and thereby prevent ß-arrestin/AP2 interaction and ß-arrestin-mediated GPCR endocytosis. In agreement with this, AP2/ß-arrestin interaction induced by an FPR2-specific agonist was inhibited by Barbadin. Unexpectedly, however, Barbadin did not inhibit FPR2 endocytosis, indicating that a mechanism independent of ß-arrestin/AP2 interaction may sustain FPR2 endocytosis. This was confirmed by the fact, that FPR2 also underwent agonist-promoted endocytosis in ß-arrestin deficient cells, albeit at a diminished level as compared to wild type cells. Dissection of the Barbadin effects on FPR2-mediated neutrophil functions including NADPH-oxidase activation mediated release of reactive oxygen species (ROS) and chemotaxis revealed that Barbadin had no effect on chemotactic migration whereas the release of ROS was potentiated/primed. The effect of Barbadin on ROS production was reversible, independent of ß-arrestin recruitment, and similar to that induced by latrunculin A. Taken together, our data demonstrate that endocytic uptake of FPR2 occurs independently of ß-arrestin, while Barbadin selectively augments FPR2-mediated ROS production independently of receptor endocytosis. Given that Barbadin binds to AP2 and prevents the AP2/ß-arrestin interaction, our results indicate a role for AP2 in FPR2-mediated ROS release from neutrophils.


Subject(s)
Endocytosis/genetics , Pyrimidines/pharmacology , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , beta-Arrestin 1/genetics , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Clathrin/chemistry , Endocytosis/drug effects , HEK293 Cells , Humans , NADPH Oxidases/genetics , Neutrophils/drug effects , Protein Binding/drug effects , Pyrimidines/chemistry , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Lipoxin/chemistry , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
11.
FASEB J ; 34(10): 13993-14005, 2020 10.
Article in English | MEDLINE | ID: mdl-32856352

ABSTRACT

Many factors are involved in the process of nerve regeneration. Understanding the mechanisms regarding how these factors promote an efficient remyelination is crucial to deciphering the molecular and cellular processes required to promote nerve repair. Schwann cells (SCs) play a central role in the process of peripheral nerve repair/regeneration. Using a model of facial nerve crush injury and repair, we identified Annexin A1 (ANXA1) as the extracellular trigger of SC proliferation and migration. ANXA1 activated formyl peptide receptor 2 (FPR2) receptors and the downstream adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling cascade, leading to SC proliferation and migration in vitro. SCs lacking FPR2 or AMPK displayed a defect in proliferation and migration. After facial nerve injury (FNI), ANXA1 promoted the proliferation of SCs and nerve regeneration in vivo. Collectively, these data identified the ANXA1/FPR2/AMPK axis as an important pathway in SC proliferation and migration. ANXA1-induced remyelination and SC proliferation promotes FNI regeneration.


Subject(s)
Annexin A1/metabolism , Cell Movement , Cell Proliferation , Facial Nerve Injuries/metabolism , Nerve Regeneration , Schwann Cells/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Annexin A1/genetics , Cells, Cultured , Male , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Schwann Cells/physiology , Signal Transduction
12.
Pharmacol Res ; 161: 105117, 2020 11.
Article in English | MEDLINE | ID: mdl-32768626

ABSTRACT

BACKGROUND AND PURPOSE: Formyl peptide receptor 2 (FPR2) is a Class A G protein-coupled receptor (GPCR) that interacts with multiple ligands and transduces both proinflammatory and anti-inflammatory signals. These ligands include weak agonists and modulators that are produced during inflammation. The present study investigates how prolonged exposure to FPR2 modulators influence receptor signaling. EXPERIMENTAL APPROACH: Fluorescent biosensors of FPR2 were constructed based on single-molecule fluorescent resonance energy transfer (FRET) and used for measurement of ligand-induced receptor conformational changes. These changes were combined with FPR2-mediated signaling events and used as parameters for the conformational states of FPR2. Ternary complex models were developed to interpret ligand concentration-dependent changes in FPR2 conformational states. KEY RESULTS: Incubation with Ac2-26, an anti-inflammatory ligand of FPR2, decreased FRET intensity at picomolar concentrations. In comparison, WKYMVm (W-pep) and Aß42, both proinflammatory agonists of FPR2, increased FRET intensity. Preincubation with Ac2-26 at 10 pM diminished W-pep-induced Ca2+ flux but potentiated W-pep-stimulated ß-arrestin2 membrane translocation and p38 MAPK phosphorylation. The opposite effects were observed with 10 pM of Aß42. Neither Ac2-26 nor Aß42 competed for W-pep binding at the picomolar concentrations. CONCLUSIONS AND IMPLICATIONS: The results support the presence of two allosteric binding sites on FPR2, each for Ac2-26 and Aß42, with high and low affinities. Sequential binding of the two allosteric ligands at increasing concentrations induce different conformational changes in FPR2, providing a novel mechanism by which biased allosteric modulators alter receptor conformations and generate pro- and anti-inflammatory signals.


Subject(s)
Amyloid beta-Peptides/pharmacology , Annexin A1/pharmacology , Inflammation Mediators/agonists , Peptide Fragments/pharmacology , Peptides/pharmacology , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Biosensing Techniques , Calcium Signaling , Cell Line, Tumor , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Ligands , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Phosphorylation , Protein Conformation , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Recombinant Fusion Proteins , Structure-Activity Relationship , beta-Arrestin 2/metabolism
13.
Front Immunol ; 11: 843, 2020.
Article in English | MEDLINE | ID: mdl-32477346

ABSTRACT

Infection, sterile injury, and chronic inflammation trigger the acute phase response in order to re-establish homeostasis. This response includes production of positive acute phase proteins in the liver, such as members of the serum amyloid A (SAA) family. In humans the major acute phase SAAs comprise a group of closely related variants of SAA1 and SAA2. SAA1 was proven to be chemotactic for several leukocyte subtypes through activation of the G protein-coupled receptor FPRL1/FPR2. Several other biological activities of SAA1, such as cytokine induction, reported to be mediated via TLRs, have been debated recently. Especially commercial SAA1, recombinantly produced in Escherichia coli, was found to be contaminated with bacterial products confounding biological assays performed with this rSAA1. We purified rSAA1 by RP-HPLC to homogeneity, removing contaminants such as lipopolysaccharides, lipoproteins and formylated peptides, and re-assessed several biological activities attributed to SAA1 (chemotaxis, cytokine induction, MMP-9 release, ROS generation, and macrophage differentiation). The homogeneous rSAA1 (hrSAA1) lacked most cell-activating properties, but its leukocyte-recruiting capacity in vivo and it's in vitro synergy with other leukocyte attractants remained preserved. Furthermore, hrSAA1 maintained the ability to promote monocyte survival. This indicates that pure hrSAA1 retains its potential to activate FPR2, whereas TLR-mediated effects seem to be related to traces of bacterial TLR ligands in the E. coli-produced human rSAA1.


Subject(s)
Leukocytes/drug effects , Leukocytes/immunology , Serum Amyloid A Protein/pharmacology , Blood Donors , Cell Differentiation/drug effects , Cell Survival/drug effects , Chemotaxis/drug effects , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , HEK293 Cells , Humans , Macrophages/drug effects , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Monocytes/drug effects , Monocytes/metabolism , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/isolation & purification , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Transfection
14.
Proc Natl Acad Sci U S A ; 117(25): 14354-14364, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513697

ABSTRACT

Ischemic injury initiates a sterile inflammatory response that ultimately participates in the repair and recovery of tissue perfusion. Macrophages are required for perfusion recovery during ischemia, in part because they produce growth factors that aid in vascular remodeling. The input signals governing this pro-revascularization phenotype remain of interest. Here we found that hindlimb ischemia increases levels of resolvin D1 (RvD1), an inflammation-resolving lipid mediator that targets macrophages via its receptor, ALX/FPR2. Exogenous RvD1 enhances perfusion recovery during ischemia, and mice deficient in Alx/Fpr2 have an endogenous defect in this process. Mechanistically, RNA sequencing revealed that RvD1 induces a transcriptional program in macrophages characteristic of a pro-revascularization phenotype. Vascularization of ischemic skeletal muscle, as well as cutaneous wounds, is impaired in mice with myeloid-specific deficiency of Alx/Fpr2, and this is associated with altered expression of pro-revascularization genes in skeletal muscle and macrophages isolated from skeletal muscle. Collectively, these results uncover a role of ALX/FPR2 in revascularization that may be amenable to therapeutic targeting in diseases associated with altered tissue perfusion and repair.


Subject(s)
Docosahexaenoic Acids/metabolism , Ischemia/immunology , Neovascularization, Physiologic/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Wound Healing/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Ischemia/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/blood supply , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Primary Cell Culture , RNA-Seq , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Signal Transduction/immunology , Skin/blood supply , Skin/immunology , Skin/injuries , Skin/pathology , Transcription, Genetic/immunology
15.
J Med Chem ; 63(17): 9003-9019, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32407089

ABSTRACT

Formyl peptide receptor 2 (FPR2) agonists can stimulate resolution of inflammation and may have utility for treatment of diseases caused by chronic inflammation, including heart failure. We report the discovery of a potent and selective FPR2 agonist and its evaluation in a mouse heart failure model. A simple linear urea with moderate agonist activity served as the starting point for optimization. Introduction of a pyrrolidinone core accessed a rigid conformation that produced potent FPR2 and FPR1 agonists. Optimization of lactam substituents led to the discovery of the FPR2 selective agonist 13c, BMS-986235/LAR-1219. In cellular assays 13c inhibited neutrophil chemotaxis and stimulated macrophage phagocytosis, key end points to promote resolution of inflammation. Cardiac structure and functional improvements were observed in a mouse heart failure model following treatment with BMS-986235/LAR-1219.


Subject(s)
Pyrrolidinones/chemistry , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Animals , Chemotaxis/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , HEK293 Cells , Heart Failure/pathology , Heart Failure/prevention & control , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Microsomes, Liver/metabolism , Neutrophils/cytology , Neutrophils/physiology , Phagocytosis/drug effects , Pyrrolidinones/metabolism , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Structure-Activity Relationship
16.
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
17.
Nat Commun ; 11(1): 885, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060286

ABSTRACT

Formylpeptide receptors (FPRs) as G protein-coupled receptors (GPCRs) can recognize formylpeptides derived from pathogens or host cells to function in host defense and cell clearance. In addition, FPRs, especially FPR2, can also recognize other ligands with a large chemical diversity generated at different stages of inflammation to either promote or resolve inflammation in order to maintain a balanced inflammatory response. The mechanism underlying promiscuous ligand recognition and activation of FPRs is not clear. Here we report a cryo-EM structure of FPR2-Gi signaling complex with a peptide agonist. The structure reveals a widely open extracellular region with an amphiphilic environment for ligand binding. Together with computational docking and simulation, the structure suggests a molecular basis for the recognition of formylpeptides and a potential mechanism of receptor activation, and reveals conserved and divergent features in Gi coupling. Our results provide a basis for understanding the molecular mechanism of the functional promiscuity of FPRs.


Subject(s)
Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/chemistry , Receptors, Lipoxin/metabolism , Animals , Binding Sites , Cryoelectron Microscopy , Humans , Ligands , Molecular Docking Simulation , Mutation , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Rats , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Signal Transduction
18.
Cytokine Growth Factor Rev ; 50: 29-42, 2019 12.
Article in English | MEDLINE | ID: mdl-31718982

ABSTRACT

Expression of the acute phase protein serum amyloid A (SAA) is dependent on the release of the pro-inflammatory cytokines IL-1, IL-6 and TNF-α during infection and inflammation. Hepatitis C virus (HCV) upregulates SAA-inducing cytokines. In line with this, a segment of chronically infected individuals display increased circulating levels of SAA. SAA has even been proposed to be a potential biomarker to evaluate treatment efficiency and the course of disease. SAA possesses antiviral activity against HCV via direct interaction with the viral particle, but might also divert infectivity through its function as an apolipoprotein. On the other hand, SAA shares inflammatory and angiogenic activity with chemotactic cytokines by activating the G protein-coupled receptor, formyl peptide receptor 2. These latter properties might promote chronic inflammation and hepatic injury. Indeed, up to 80 % of infected individuals develop chronic disease because they cannot completely clear the infection, due to diversion of the immune response. In this review, we summarize the interconnection between SAA and cytokines in the context of HCV infection and highlight the dual role SAA could play in this disease. Nevertheless, more research is needed to establish whether the balance between those opposing activities can be tilted in favor of the host defense.


Subject(s)
Cytokines/immunology , Hepatitis C/immunology , Hepatitis C/physiopathology , Serum Amyloid A Protein/immunology , Animals , Cytokines/genetics , Hepacivirus , Humans , Inflammation , Liver/immunology , Liver/pathology , Mice , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/genetics , Receptors, Lipoxin/immunology , Serum Amyloid A Protein/analysis
19.
J Exp Clin Cancer Res ; 38(1): 170, 2019 Apr 18.
Article in English | MEDLINE | ID: mdl-30999932

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) require stromal signals for maintaining pluripotency and self-renewal capacities to confer tumor metastasis. Resolvin D1 (RvD1), an endogenous anti-inflammatory lipid mediator, has recently been identified to display anti-cancer effects by acting on stroma cells. Our previous study reveals that hepatic stellate cells (HSCs)-derived cartilage oligomeric matrix protein (COMP) contributes to hepatocellular carcinoma (HCC) progression. However, whether RvD1 inhibits paracrine of cancer-associated fibroblasts (CAFs)-derived COMP to prevent epithelial-mesenchymal transition (EMT) and cancer stemness in HCC remains to be elucidated. METHODS: CAFs were isolated from HCC tissues. Direct and indirect co-culture models were established to analyze the interactions between HCC cells and CAFs in the presence of RvD1 in vitro. The transwell and tumor sphere formation assays were used to determine invasion and stemness of HCC cells. The subcutaneous tumor formation and orthotopic liver tumor models were established by co-implantation of CAFs and HCC cells to evaluate the role of RvD1 in vivo. To characterize the mechanism of RvD1 inhibited paracrine of COMP in CAFs, various signaling molecules were analyzed by ELISA, western blotting, reactive oxygen species (ROS) detection, immunofluorescence staining, dual luciferase reporter assay and chromatin immunoprecipitation assay. RESULTS: Our data revealed that RvD1 treatment can impede the CAFs-induced cancer stem-like properties and the EMT of HCC cells under co-culture conditions. In vivo studies indicated that RvD1 intervention repressed the promoting effects of CAFs on tumor growth and metastasis of HCC. Furthermore, RvD1 inhibited CAF-induced EMT and stemness features of HCC cells by suppressing the secretion of COMP. Mechanistically, formyl peptide receptor 2 (FPR2) receptor mediated the suppressive effects of RvD1 on COMP and forkhead box M1 (FOXM1) expression in CAFs. Notably, RvD1 impaired CAF-derived COMP in a paracrine manner by targeting FPR2/ROS/FOXM1 signaling to ultimately abrogate FOXM1 recruitment to the COMP promoter. CONCLUSION: Our results indicated that RvD1 impaired paracrine of CAFs-derived COMP by targeting FPR2/ROS/FOXM1 signaling to repress EMT and cancer stemness in HCC. Thus, RvD1 may be a potential agent to promote treatment outcomes in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cartilage Oligomeric Matrix Protein/genetics , Forkhead Box Protein M1/genetics , Liver Neoplasms/drug therapy , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Docosahexaenoic Acids , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Paracrine Communication/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
20.
Am J Reprod Immunol ; 81(5): e13107, 2019 05.
Article in English | MEDLINE | ID: mdl-30811719

ABSTRACT

PROBLEM: To test whether lipoxin A4 (LXA4) interferes with embryo implantation via suppression of epithelial-mesenchymal transition (EMT). METHOD OF STUDY: We developed a mouse model of LXA4 blocking embryo implantation and detected the indicators of EMT to confirm that LXA4 inhibits EMT might be a mechanism of interfering with the embryo implantation. We detected integrin-linked kinase (ILK), N-formylpeptide receptor 2 (FPR2), vascular endothelial growth factor, matrix metalloproteinases (MMPs), Akt, GSK3ß, NF-ĸB, twist, vimentin, fibronectin, and ß-catenin mRNA expression using reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time RT-PCR; localized protein expression using immunohistochemistry and Western blotting assay; MMPs activity assay by gelatin zymography; and the status of implantation in pregnant animals assessed by pontamine blue reaction test. RESULTS: Preimplantation administration of LXA4 resulted in implantation failure. LXA4 has a time- and dose-dependent effect on embryo implantation. Day 0.5 after fertilization is the most effective time to use LXA4 to block embryo implantation. (a) LXA4 reduced endometrial stroma edema; (b) LXA4 inhibited the activity of MMP9 and significantly upregulated the expression of ß-catenin, and downregulated the expression of vimentin, fibronectin, twist, NF-κB, Akt, and Gsk-3ß in the endometrium and TEV-1 cells; (c) LXA4 upregulated the expression of FPR2, and downregulated the expression of ILK; FPR2-overexpressing had an inhibitory effect on ILK in TEV-1 cells. CONCLUSION: LXA4 inhibits EMT which attenuates ILK action by enhancing FPR2; therefore, this might be a mechanism of interfering with embryo implantation.


Subject(s)
Infertility, Female/immunology , Lipoxins/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Animals , Cells, Cultured , Embryo Implantation , Epithelial-Mesenchymal Transition , Female , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Models, Animal , Pregnancy , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...