Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 14: 1119227, 2023.
Article in English | MEDLINE | ID: mdl-36817589

ABSTRACT

Originally, it was thought that a single serum amyloid A (SAA) protein was involved in amyloid A amyloidosis, but in fact, SAA represents a four-membered family wherein SAA1 and SAA2 are acute phase proteins (A-SAA). SAA is highly conserved throughout evolution within a wide range of animal species suggestive of an important biological function. In fact, A-SAA has been linked to a number of divergent biological activities wherein a number of these functions are mediated via the G protein-coupled receptor (GPCR), formyl peptide receptor (FPR) 2. For instance, through the activation of FPR2, A-SAA has been described to regulate leukocyte activation, atherosclerosis, pathogen recognition, bone formation and cell survival. Moreover, A-SAA is subject to post-translational modification, primarily through proteolytic processing, generating a range of A-SAA-derived peptides. Although very little is known regarding the biological effect of A-SAA-derived peptides, they have been shown to promote neutrophil and monocyte migration through FPR2 activation via synergy with other GPCR ligands namely, the chemokines CXCL8 and CCL3, respectively. Within this review, we provide a detailed analysis of the FPR2-mediated functions of A-SAA. Moreover, we discuss the potential role of A-SAA-derived peptides as allosteric modulators of FPR2.


Subject(s)
Receptors, Formyl Peptide , Serum Amyloid A Protein , Animals , Receptors, Formyl Peptide/physiology , Ligands , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/pharmacology , Signal Transduction , Peptides/metabolism
2.
Semin Immunol ; 59: 101602, 2022 01.
Article in English | MEDLINE | ID: mdl-35277300

ABSTRACT

Formyl peptide receptor type 2 (FPR2) regulates the initiation and resolution phases of the inflammatory response. In the setting of heart injury and disease, dysregulated inflammation can potentiate maladaptive healing and pathological remodeling of the heart leading to cardiac dysfunction and failure. The potential to regulate and resolve adverse inflammation is postulated to improve outcome in the setting of heart disease. This review covers emerging concepts on the role of FPR2 in heart disease and strategies to activate pro-resolution processes to limit disease progression. We summarize key preclinical studies that support use of FPR2 agonists in heart disease. Finally, we briefly discuss the status of FPR2 agonists under evaluation in the clinic.


Subject(s)
Heart Diseases , Receptors, Formyl Peptide , Humans , Inflammation/pathology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/physiology , Wound Healing
3.
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
4.
Mol Med Rep ; 24(6)2021 12.
Article in English | MEDLINE | ID: mdl-34608500

ABSTRACT

Recurrent spontaneous abortion (RSA) effects both the physical and mental health of women of reproductive age. Trophoblast dysfunction may result in RSA due to shallow placental implantation. The mechanisms underlying formyl peptide receptor 2 (FPR2) on the biological functions of trophoblasts remain to be elucidated. The present study aimed to explore the potential functions of FPR2, a G protein­coupled receptor, in placental trophoblasts. The location and expression levels of FPR2 in the villi tissue of patients with RSA were detected using immunohistochemical staining, reverse transcription­quantitative PCR and western blotting. Following the transfection of small interfering RNA targeting FPR2 in HTR­8/SVneo cells, a Cell Counting Kit­8 assay was used to determine the levels of cell viability. Flow cytometry was used to examine the levels of cell apoptosis and gap closure and Transwell assays were carried out to evaluate the levels of cell migration and invasion. A tube formation assay was performed to detect the levels of capillary­like structure formation. Western blotting was used to detect the expression levels of proteins in the associated signaling pathways. The expression of FPR2 was present in villi trophoblasts and was markedly increased in patients with RSA. The levels of trophoblast invasion, migration and tube formation were markedly increased following FPR2 knockdown, whereas the levels of apoptosis were markedly decreased. In addition, FPR2 knockdown caused an increase in the phosphorylation levels of AKT and PI3K. Thus, FPR2 may be involved in the regulation of trophoblast function via the PI3K/AKT signaling pathway. The results of the present study provided a theoretical basis for the use of FPR2 as a target for the treatment of trophoblast­associated diseases, such as RSA.


Subject(s)
Abortion, Habitual/physiopathology , Chorionic Villi/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Trophoblasts/metabolism , Apoptosis , Cell Line , Cell Movement , Cell Survival , Female , Gene Knockdown Techniques/methods , Humans , Pregnancy , RNA, Small Interfering/metabolism , Signal Transduction
5.
Cell Death Dis ; 11(8): 653, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32811815

ABSTRACT

Radiation therapy is an important modality in the treatment of lung cancer, but it can lead to radiation pneumonitis, and eventually radiation fibrosis. To date, only few available drugs can effectively manage radiation-induced pulmonary fibrosis. Lipoxins are endogenous molecules exhibit anti-inflammatory and pro-resolving effects. These molecules play a vital role in reducing excessive tissue injury and chronic inflammation; however, their effects on radiation-induced lung injury (RILI) are unknown. In this study, we investigated the effects of lipoxin A4 (LXA4) on RILI using our specialized small-animal model of RILI following focal-ablative lung irradiation (IR). LXA4 significantly inhibited immune-cell recruitment and reduced IR-induced expression of pro-inflammatory cytokines and fibrotic proteins in the lung lesion sites. In addition, micro-CT revealed that LXA4 reduced IR-induced increases in lung consolidation volume. The flexiVentTM assays showed that LXA4 significantly reversed IR-induced lung function damage. Moreover, LXA4 downregulated the activities of NF-κB and the Smad-binding element promoters. The expression of FPR2, an LXA4 receptor, increased during the development of IR-induced pulmonary fibrosis, whereas silencing of endogenous LXA4 using an antagonist (WRW4) or FPR2 siRNA resulted in impaired development of pulmonary fibrosis in response to IR. Collectively, these data suggest that LXA4 could serve as a potent therapeutic agent for alleviating RILI.


Subject(s)
Lipoxins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Fibrosis/metabolism , Humans , Lipoxins/physiology , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/physiopathology , Radiation , Radiotherapy/adverse effects , Receptor Cross-Talk/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/metabolism , Receptors, Lipoxin/physiology , Signal Transduction/physiology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
6.
Article in English | MEDLINE | ID: mdl-32038501

ABSTRACT

A hallmark of inflammatory responses is leukocyte mobilization, which is mediated by pathogen and host released chemotactic factors that activate Gi-protein-coupled seven-transmembrane receptors (GPCRs) on host cell surface. Formylpeptide receptors (FPRs, Fprs in mice) are members of the chemoattractant GPCR family, shown to be critical in myeloid cell trafficking during infection, inflammation, immune responses, and cancer progression. Accumulating evidence demonstrates that both human FPRs and murine Fprs are involved in a number of patho-physiological processes because of their expression on a wide variety of cell types in addition to myeloid cells. The unique capacity of FPRs (Fprs) to interact with numerous structurally unrelated chemotactic ligands enables these receptors to participate in orchestrated disease initiation, progression, and resolution. One murine Fpr member, Fpr2, and its endogenous agonist peptide, Cathelicidin-related antimicrobial peptide (CRAMP), have been demonstrated as key mediators of colon mucosal homeostasis and protection from inflammation and associated tumorigenesis. Recent availability of genetically engineered mouse models greatly expanded the understanding of the role of FPRs (Fprs) in pathophysiology that places these molecules in the list of potential targets for therapeutic intervention of diseases.


Subject(s)
Chemotactic Factors/physiology , Inflammation/etiology , Neoplasms/etiology , Receptors, Formyl Peptide/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Homeostasis/genetics , Humans , Inflammation/genetics , Ligands , Mice , Neoplasms/genetics
7.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-32102985

ABSTRACT

Neutrophils are the most abundant inflammatory cells at the earliest stages of wound healing and play important roles in wound repair and fibrosis. Formyl peptide receptor 1 (FPR-1) is abundantly expressed on neutrophils and has been shown to regulate their function, yet the importance of FPR-1 in fibrosis remains ill defined. FPR-1-deficient (fpr1-/-) mice were protected from bleomycin-induced pulmonary fibrosis but developed renal and hepatic fibrosis normally. Mechanistically, we observed a failure to effectively recruit neutrophils to the lungs of fpr1-/- mice, whereas neutrophil recruitment was unaffected in the liver and kidney. Using an adoptive transfer model we demonstrated that the defect in neutrophil recruitment to the lung was intrinsic to the fpr1-/- neutrophils, as C57BL/6 neutrophils were recruited normally to the damaged lung in fpr1-/- mice. Finally, C57BL/6 mice in which neutrophils had been depleted were protected from pulmonary fibrosis. In conclusion, FPR-1 and FPR-1 ligands are required for effective neutrophil recruitment to the damaged lung. Failure to recruit neutrophils or depletion of neutrophils protects from pulmonary fibrosis.


Subject(s)
Neutrophil Infiltration/physiology , Pulmonary Fibrosis/physiopathology , Receptors, Formyl Peptide/physiology , Animals , Bleomycin/toxicity , Humans , Ligands , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism
8.
FASEB J ; 33(12): 13632-13643, 2019 12.
Article in English | MEDLINE | ID: mdl-31585047

ABSTRACT

Mucosal wound repair is coordinated by dynamic crosstalk between endogenous and exogenous mediators and specific receptors on epithelial cells and infiltrating immune cells. One class of such receptor-ligand pairs involves formyl peptide receptors (FPRs) that have been shown to influence inflammatory response and repair. Here we explored the role of murine Fpr2/3, an ortholog of human FPR2/receptor for lipoxin A4 (ALX), in orchestrating intestinal mucosal repair. Compared with wild-type (WT) mice, Fpr2/3-/- mice exhibited delayed recovery from acute experimental colitis and perturbed repair after biopsy-induced colonic mucosal injury. Decreased numbers of infiltrating monocytes were observed in healing wounds from Fpr2/3-/- mice compared with WT animals. Bone marrow transplant experiments revealed that Fpr2/3-/- monocytes showed a competitive disadvantage when infiltrating colonic wounds. Moreover, Fpr2/3-/- monocytes were defective in chemotactic responses to the chemokine CC chemokine ligand (CCL)20, which is up-regulated during early phases of inflammation. Analysis of Fpr2/3-/- monocytes revealed altered expression of the CCL20 receptor CC chemokine receptor (CCR)6, suggesting that Fpr2/3 regulates CCL20-CCR6-mediated monocyte chemotaxis to sites of mucosal injury in the gut. These findings demonstrate an important contribution of Fpr2/3 in facilitating monocyte recruitment to sites of mucosal injury to influence wound repair.-Birkl, D., O'Leary, M. N., Quiros, M., Azcutia, V., Schaller, M., Reed, M., Nishio, H., Keeney, J., Neish, A. S., Lukacs, N. W., Parkos, C. A., Nusrat, A. Formyl peptide receptor 2 regulates monocyte recruitment to promote intestinal mucosal wound repair.


Subject(s)
Cell Movement , Inflammation/therapy , Intestinal Mucosa/physiology , Monocytes/metabolism , Receptors, Formyl Peptide/physiology , Wound Healing , Animals , Bone Marrow Transplantation , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/toxicity , Inflammation/etiology , Inflammation/pathology , Intestinal Mucosa/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Receptors, CCR6/genetics , Receptors, CCR6/metabolism
9.
PLoS Biol ; 17(8): e3000395, 2019 08.
Article in English | MEDLINE | ID: mdl-31465435

ABSTRACT

The gastric pathogen Helicobacter pylori requires a noncanonical cytosolic chemoreceptor transducer-like protein D (TlpD) for efficient colonization of the mammalian stomach. Here, we reconstituted a complete chemotransduction signaling complex in vitro with TlpD and the chemotaxis (Che) proteins CheW and CheA, enabling quantitative assays for potential chemotaxis ligands. We found that TlpD is selectively sensitive at micromolar concentrations to bleach (hypochlorous acid, HOCl), a potent antimicrobial produced by neutrophil myeloperoxidase during inflammation. HOCl acts as a chemoattractant by reversibly oxidizing a conserved cysteine within a 3His/1Cys Zn-binding motif in TlpD that inactivates the chemotransduction signaling complex. We found that H. pylori is resistant to killing by millimolar concentrations of HOCl and responds to HOCl in the micromolar range by increasing its smooth-swimming behavior, leading to chemoattraction to HOCl sources. We show related protein domains from Salmonella enterica and Escherichia coli possess similar reactivity toward HOCl. We propose that this family of proteins enables host-associated bacteria to sense sites of tissue inflammation, a strategy that H. pylori uses to aid in colonizing and persisting in inflamed gastric tissue.


Subject(s)
Chemotaxis/physiology , Helicobacter pylori/metabolism , Receptors, Formyl Peptide/metabolism , Bacterial Proteins/metabolism , Bleaching Agents , Chemoreceptor Cells/metabolism , Chemotactic Factors/metabolism , Cytosol/metabolism , Cytosol/physiology , Helicobacter pylori/physiology , Hypochlorous Acid , Oxidation-Reduction , Receptors, Formyl Peptide/physiology , Signal Transduction
10.
FASEB J ; 33(11): 12188-12199, 2019 11.
Article in English | MEDLINE | ID: mdl-31398292

ABSTRACT

Pattern recognition receptors (PRRs) are key elements in the innate immune response. Formyl peptide receptor (FPR) 2 is a PRR that, in addition to proinflammatory, pathogen-derived compounds, also recognizes the anti-inflammatory endogenous ligand annexin A1 (AnxA1). Because the contribution of this signaling axis in viral infections is undefined, we investigated AnxA1-mediated FPR2 activation on influenza A virus (IAV) infection in the murine model. AnxA1-treated mice displayed significantly attenuated pathology upon a subsequent IAV infection with significantly improved survival, impaired viral replication in the respiratory tract, and less severe lung damage. The AnxA1-mediated protection against IAV infection was not caused by priming of the type I IFN response but was associated with an increase in the number of alveolar macrophages (AMs) and enhanced pulmonary expression of the AM-regulating cytokine granulocyte-M-CSF (GM-CSF). Both AnxA1-mediated increase in AM levels and GM-CSF production were abrogated when mouse (m)FPR2 signaling was antagonized but remained up-regulated in mice genetically deleted for mFPR1, an mFPR2 isoform also serving as AnxA1 receptor. Our results indicate a novel protective function of the AnxA1-FPR2 signaling axis in IAV pathology via GM-CSF-associated maintenance of AMs, expanding knowledge on the potential use of proresolving mediators in host defense against pathogens.-Schloer, S., Hübel, N., Masemann, D., Pajonczyk, D., Brunotte, L., Ehrhardt, C., Brandenburg, L.-O., Ludwig, S., Gerke, V., Rescher, U. The annexin A1/FPR2 signaling axis expands alveolar macrophages, limits viral replication, and attenuates pathogenesis in the murine influenza A virus infection model.


Subject(s)
Annexin A1/physiology , Influenza A virus/physiology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/prevention & control , Receptors, Formyl Peptide/physiology , Virus Replication , Animals , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Influenza A virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
11.
J Vis Exp ; (147)2019 05 24.
Article in English | MEDLINE | ID: mdl-31180367

ABSTRACT

Various limitations of 2D cell culture systems have sparked interest in 3D cell culture and analysis platforms, which would better mimic the spatial and chemical complexity of living tissues and mimic in vivo tissue functions. Recent advances in microfabrication technologies have facilitated the development of 3D in vitro environments in which cells can be integrated into a well-defined extracellular matrix (ECM) and a defined set of soluble or matrix associated biomolecules. However, technological barriers have limited their widespread use in research laboratories. Here, we describe a method to construct simple devices for 3D culture and experimentation with cells and multicellular organoids in 3D microenvironments with a defined chemoattractant gradient. We illustrate the use of this platform for analysis of the response of epithelial cells and organoids to gradients of growth factors, such as epidermal growth factor (EGF). EGF gradients were stable in the devices for several days leading to directed branch formation in breast organoids. This analysis allowed us to conclude that collective gradient sensing by groups of cells is more sensitive vs. single cells. We also describe the fabrication method, which does not require photolithography facilities nor advanced soft lithography techniques. This method will be helpful to study 3D cellular behaviors in the context of the analysis of development and pathological states, including cancer.


Subject(s)
Extracellular Matrix/metabolism , Imaging, Three-Dimensional/methods , Receptors, Formyl Peptide/physiology , Animals , Humans , Mice
12.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 305-316, 2019 02.
Article in English | MEDLINE | ID: mdl-30521870

ABSTRACT

The innate immune system is the first line of defense against pathogenic threats. For the early pathogen recognition and activation of cell protective mechanisms, germline-encoded pattern recognition receptors (PRRs) detect characteristic and evolutionary conserved pathogen-associated molecular patterns (PAMPs). PRRs are therefore key elements in the innate immune response; in addition, they sense danger-associated molecular patterns (DAMPs) that are released by host cell molecules under pathophysiological conditions. Formyl peptide receptors (FPRs) are G-protein-coupled PRRs that respond to a surprisingly broad range of ligands, derived from both pathogens and host cells. Here, we exemplary discuss ligands in order to illustrate the wide pathophysiological relevance of the FPR signaling axis in case of e.g., chronic inflammations and to underscore its potential therapeutic value in the light of "biased agonism", a modern concept of GPCR (G-protein coupled receptors) activation. These novel insights into the GPCR receptor biochemistry will hopefully (re)stimulate FPR-related research and lead to novel strategies for the urgently needed development of drugs with pharmacologically advantageous characteristics.


Subject(s)
Receptors, Formyl Peptide/metabolism , Receptors, Formyl Peptide/physiology , Alarmins/metabolism , Animals , Humans , Immune System/immunology , Immunity, Innate/immunology , Inflammation/immunology , Ligands , Mice , Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction
13.
BMB Rep ; 51(8): 418-423, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30021674

ABSTRACT

Emergency granulopoiesis is a very important strategy to supply efficient neutrophil number in response to infection. However, molecular mechanism involved in this process remains unclear. Here, we found that administration of WKYMVm, an immune modulating peptide, to septic mice strongly increased neutrophil number through augmented emergency granulopoiesis. WKYMVm-induced emergency granulopoiesis was blocked not only by a formyl peptide receptor 2 (FPR2) antagonist (WRW4), but also by FPR2 deficiency. As progenitors of neutrophils, Lin-c-kit+Sca-1- cells expressed FPR2. WKYMVm-induced emergency granulopoiesis was also blocked by a phospholipase C inhibitor (U-73122). These results suggest that WKYMVm can stimulate emergency granulopoiesis via FPR2 and phospholipase C enzymatic activity. [BMB Reports 2018; 51(8): 418-423].


Subject(s)
Hematopoiesis/drug effects , Neutrophils/drug effects , Oligopeptides/pharmacology , Receptors, Formyl Peptide/metabolism , Type C Phospholipases/metabolism , Animals , Drug Interactions , Estrenes/pharmacology , Male , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/enzymology , Neutrophils/metabolism , Pyrrolidinones/pharmacology , Receptors, Formyl Peptide/physiology , Signal Transduction/physiology , Type C Phospholipases/antagonists & inhibitors
14.
Am J Pathol ; 188(10): 2195-2206, 2018 10.
Article in English | MEDLINE | ID: mdl-30031729

ABSTRACT

The most important risk factor for chronic obstructive pulmonary disease (COPD) is cigarette smoking. Until now, smoking cessation (SC) is the only treatment effective in slowing down the progression of the disease. However, in many cases SC may only relieve the airflow obstruction and inflammatory response. Consequently, a persistent lung inflammation in ex-smokers is associated with progressive deterioration of respiratory functions. This is an increasingly important clinical problem whose mechanistic basis remains poorly understood. Available therapies do not adequately suppress inflammation and are not able to stop the vicious cycle that is at the basis of persistent inflammation. In addition, in mice after SC an ongoing inflammation and progressive lung deterioration is observed. After 4 months of smoke exposure mice show mild emphysematous changes. Lung inflammation is still present after SC, and emphysema progresses during the next 6-month period of observation. Destruction of alveolar walls is associated with airways remodeling (goblet cell metaplasia and peribronchiolar fibrosis). Modulation of formyl-peptide receptor signaling with antagonists mitigates inflammation and prevents deterioration of lung structures. This study suggests an important role for N-formylated peptides in the progression and exacerbation of COPD. Modulating formyl-peptide receptor signal should be explored as a potential new therapy for COPD.


Subject(s)
Cigarette Smoking/adverse effects , Pneumonia/physiopathology , Receptors, Formyl Peptide/antagonists & inhibitors , Smoking Cessation , Airway Remodeling/physiology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cigarette Smoking/physiopathology , Disease Progression , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/physiopathology , Receptors, Formyl Peptide/physiology
15.
Am J Pathol ; 188(7): 1555-1562, 2018 07.
Article in English | MEDLINE | ID: mdl-29684359

ABSTRACT

Lipoxin receptor (ALX)/N-formyl peptide receptor (FPR)-2 is a G-protein-coupled receptor that has multiple binding partners, including the endogenous lipid mediators resolvin D1, lipoxin A4, and the Ca2+-dependent phospholipid-binding protein annexin A1. Previous studies have demonstrated that resolvin D1 activates ALX/Fpr2 to resolve salivary gland inflammation in the NOD/ShiLtJ mouse model of Sjögren syndrome. Moreover, mice lacking the ALX/Fpr2 display an exacerbated salivary gland inflammation in response to lipopolysaccharide. Additionally, activation of ALX/Fpr2 has been shown to be important for regulating antibody production in B cells. These previous studies indicate that ALX/Fpr2 promotes resolution of salivary gland inflammation while modulating adaptive immunity, suggesting the need for investigation of the role of ALX/Fpr2 in regulating antibody production and secretory function in mouse salivary glands. Our results indicate that aging female knockout mice lacking ALX/Fpr2 display a significant reduction in saliva flow rates and weight loss, an increased expression of autoimmune-associated genes, an up-regulation of autoantibody production, and increased CD20-positive B-cell population. Although not all effects were noted among the male knockout mice, the results nonetheless indicate that ALX/Fpr2 is clearly involved in the adaptive immunity and secretory function in salivary glands, with further investigation warranted to determine the cause(s) of these between-sex differences.


Subject(s)
Adaptive Immunity/immunology , Homeodomain Proteins/physiology , Inflammation/immunology , Receptors, Formyl Peptide/physiology , Salivary Glands/immunology , Submandibular Gland/immunology , Animals , Female , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Salivary Glands/metabolism , Salivary Glands/pathology , Signal Transduction , Submandibular Gland/metabolism , Submandibular Gland/pathology , Weight Loss
16.
Exp Cell Res ; 367(2): 282-290, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29649428

ABSTRACT

BACKGROUND: Annexin A1 (ANXA1) and Translocator Protein-18KDa (TSPO) down-regulate neuroinflammation. We investigated the role of recombinant ANXA1 (rANXA) on TSPO functions on Toll Like Receptor (TLR) activated microglia. METHODS: BV-2 cells (murine microglia), were stimulated by E. coli Lipopolysaccharide (LPS) and treated with rANXA1 in order to measure TSPO expression and inflammatory parameters. Anti-sense ANXA1 and TLR4 and TSPO shRNA, as well as pharmacological treatments, were employed to assess the mechanisms involved. RESULTS: LPS-stimulated BV-2 cells caused overexpression of TSPO, which was inhibited by: pharmacological blockade of TLR4 or TLR4 mRNA silencing; inhibition of myeloid differentiation primary response gene 88 (MyD88) dimerization; or blocking of nuclear factor κB (NF-κB) activation. rANXA1 treatment impaired LPS-induced TSPO upregulation by down-modulating MyD88 and NF-κB signaling; the effect was abolished by WRW4, an antagonist of formyl peptide receptor 2 (FPR2). rANXA1 treatment also downregulated interleukin 1ß (IL-1ß) and tumor necrosis factor-α (TNFα) secretion in LPS-stimulated BV-2 cells. TSPO knockdown in BV-2 cells augmented LPS-induced TNFα secretion and abolished the inhibitory effect of rANXA1 on TNFα secretion evoked by LPS. CONCLUSIONS: exogenous ANXA1 down-modulates LPS-induced TSPO via MyD-88/NF-κB pathways, and constitutive TSPO is pivotal for the control of ANXA1 on TNFα secretion. TSPO actions may be involved with the mechanisms of ANXA1 on inflammatory brain diseases.


Subject(s)
Annexin A1/physiology , Receptors, GABA/metabolism , Animals , Annexin A1/metabolism , Cell Line , Cytokines/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Receptors, Formyl Peptide/physiology , Signal Transduction , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
PLoS One ; 11(10): e0164674, 2016.
Article in English | MEDLINE | ID: mdl-27792738

ABSTRACT

Eukaryotic cells respond to a chemoattractant gradient by forming intracellular gradients of signaling molecules that reflect the extracellular chemical gradient-an ability called directional sensing. Quantitative experiments have revealed two characteristic input-output relations of the system: First, in a static chemoattractant gradient, the shapes of the intracellular gradients of the signaling molecules are determined by the relative steepness, rather than the absolute concentration, of the chemoattractant gradient along the cell body. Second, upon a spatially homogeneous temporal increase in the input stimulus, the intracellular signaling molecules are transiently activated such that the response magnitudes are dependent on fold changes of the stimulus, not on absolute levels. However, the underlying mechanism that endows the system with these response properties remains elusive. Here, by adopting a widely used modeling framework of directional sensing, local excitation and global inhibition (LEGI), we propose a hypothesis that the two rescaling behaviors stem from a single design principle, namely, invariance of the governing equations to a scale transformation of the input level. Analyses of the LEGI-based model reveal that the invariance can be divided into two parts, each of which is responsible for the respective response properties. Our hypothesis leads to an experimentally testable prediction that a system with the invariance detects relative steepness even in dynamic gradient stimuli as well as in static gradients. Furthermore, we show that the relation between the response properties and the scale invariance is general in that it can be implemented by models with different network topologies.


Subject(s)
Chemotaxis/physiology , Eukaryotic Cells/physiology , Chemotactic Factors/physiology , Feedback , Models, Biological , Receptors, Formyl Peptide/physiology , Spatio-Temporal Analysis
18.
Oncotarget ; 7(34): 54474-54487, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27323409

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility and angiogenesis by interacting with the formyl peptide receptor type 1 (FPR1). In this study, we present evidence that the cyclization of the uPAR88-92 sequence generates a new potent inhibitor of migration, and extracellular matrix invasion of human osteosarcoma and chondrosarcoma cells expressing comparable levels of FPR1 on cell surface. In vitro, the cyclized peptide [SRSRY] prevents formation of capillary-like tubes by endothelial cells co-cultured with chondrosarcoma cells and trans-endothelial migration of osteosarcoma and chondrosarcoma cells. When chondrosarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density and circulating tumor cells in blood samples collected before the sacrifice, were significantly reduced in animals treated daily with i.p-administration of 6 mg/Kg [SRSRY] as compared to animals treated with vehicle only. Our findings indicate that [SRSRY] prevents three key events occurring during the metastatic process of osteosarcoma and chondrosarcoma cells: the extracellular matrix invasion, the formation of a capillary network and the entry into bloodstream.


Subject(s)
Bone Neoplasms/blood supply , Chondrosarcoma/blood supply , Neovascularization, Pathologic/drug therapy , Osteosarcoma/blood supply , Peptides, Cyclic/therapeutic use , Receptors, Urokinase Plasminogen Activator/therapeutic use , Animals , Cell Line, Tumor , Cell Movement , Chondrosarcoma/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Osteosarcoma/pathology , Receptors, Formyl Peptide/physiology
19.
Circulation ; 133(22): 2169-79, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27154726

ABSTRACT

BACKGROUND: Platelet activation at sites of vascular injury is essential for hemostasis, but it is also a major pathomechanism underlying ischemic injury. Because anti-inflammatory therapies limit thrombosis and antithrombotic therapies reduce vascular inflammation, we tested the therapeutic potential of 2 proresolving endogenous mediators, annexin A1 N-terminal derived peptide (AnxA1Ac2-26) and aspirin-triggered lipoxin A4 (15-epi-lipoxin A4), on the cerebral microcirculation after ischemia/reperfusion injury. Furthermore, we tested whether the lipoxin A4 receptor formyl-peptide receptor 2/3 (Fpr2/3; ortholog to human FPR2/lipoxin A4 receptor) evoked neuroprotective functions after cerebral ischemia/reperfusion injury. METHODS AND RESULTS: Using intravital microscopy, we found that cerebral ischemia/reperfusion injury was accompanied by neutrophil and platelet activation and neutrophil-platelet aggregate formation within cerebral microvessels. Moreover, aspirin-triggered lipoxin A4 activation of neutrophil Fpr2/3 regulated neutrophil-platelet aggregate formation in the brain and inhibited the reactivity of the cerebral microvasculature. The same results were obtained with AnxA1Ac2-26 administration. Blocking Fpr2/lipoxin A4 receptor with the antagonist Boc2 reversed this effect, and treatments were ineffective in Fpr2/3 knockout mice, which displayed an exacerbated disease severity, evidenced by increased infarct area, blood-brain barrier dysfunction, increased neurological score, and elevated levels of cytokines. Furthermore, aspirin treatment significantly reduced cerebral leukocyte recruitment and increased endogenous levels of aspirin-triggered lipoxin A4, effects again mediated by Fpr2/3. CONCLUSION: Fpr2/lipoxin A4 receptor is a therapeutic target for initiating endogenous proresolving, anti-inflammatory pathways after cerebral ischemia/reperfusion injury.


Subject(s)
Cardiovascular Diseases/therapy , Cerebral Infarction/pathology , Neutrophils/physiology , Receptors, Formyl Peptide/physiology , Amino Acid Sequence , Animals , Annexin A1/genetics , Annexin A1/pharmacology , Annexin A1/therapeutic use , Cardiovascular Diseases/pathology , Cerebral Infarction/prevention & control , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Formyl Peptide/agonists
20.
Front Biosci (Elite Ed) ; 8(3): 436-49, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27100350

ABSTRACT

Neutrophils play a critical role in acute and chronic inflammatory diseases. N-formyl peptides, which originate from bacterial peptides or mitochondrial proteins bind with a high binding affinity to formyl peptide receptor 1 (FPR1). N-formyl peptide-FPR1 is involved in the pathogenesis of sterile and infectious inflammatory processes and causes phagocytosis of pathogens or injured cells by neutrophils. Excessive activation of neutrophils by binding of N-formyl peptides is associated with tissue injury requiring drugs that block FPR1-dependent signaling. Here, we review the roles of FPR1 as a critical regulator of inflammatory processes and its involvement in pathological conditions.


Subject(s)
Inflammation/immunology , Neutrophils/immunology , Receptors, Formyl Peptide/physiology , Chemotaxis, Leukocyte , Neutrophil Activation , Neutrophil Infiltration , Phagocytosis , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...