1
|
Formyl peptide receptor 2 is an emerging modulator of inflammation in the liver. Exp Mol Med 2023; 55:325-332. [PMID: 36750693 PMCID: PMC9981720 DOI: 10.1038/s12276-023-00941-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 02/09/2023] Open
Abstract
Formyl peptide receptors (FPRs), which are seven-membrane G-protein coupled receptors, recognize chemotactic signals to protect hosts from pathogenic infections and mediate inflammatory responses in the body. There are three isoforms of FPRs in humans-FPR1, FPR2, and FPR3-and they bind to N-formyl peptides, except FPR3, and to various endogenous agonists. Among FPR family members, FPR2 has a lower affinity for N-formyl peptides than FPR1 and binds with a wide range of endogenous or exogenous agonists. Thus, FPR2 is considered the most ambiguous member. Accumulating evidence has shown that FPR2 is involved in the host's defense against bacterial infection and inflammation in liver diseases, such as nonalcoholic fatty liver disease, liver fibrosis, and liver cancer, suggesting the pathophysiological relevance of FPR2 to the liver. However, FPR2 has been shown to promote or suppress inflammation, depending on the type of FPR2-expressing cell and FPR2-bound ligands in the liver. Therefore, it is important to understand FPR2's function per se and to elucidate the mechanism underlying immunomodulation initiated by ligand-activated FPR2 before suggesting FPR2 as a novel therapeutic agent for liver diseases. In this review, up-to-date knowledge of FPR2, with general information on the FPR family, is provided. We shed light on the dual action of FPR2 in the liver and discuss the hepatoprotective roles of FPR2 itself and FPR2 agonists in mediating anti-inflammatory responses.
Collapse
|
2
|
Zhu J, Li L, Ding J, Huang J, Shao A, Tang B. The Role of Formyl Peptide Receptors in Neurological Diseases via Regulating Inflammation. Front Cell Neurosci 2021; 15:753832. [PMID: 34650406 PMCID: PMC8510628 DOI: 10.3389/fncel.2021.753832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a group of G protein-coupled cell surface receptors that play important roles in host defense and inflammation. Owing to the ubiquitous expression of FPRs throughout different cell types and since they interact with structurally diverse chemotactic agonists, they have a dual function in inflammatory processes, depending on binding with different ligands so that accelerate or inhibit key intracellular kinase-based regulatory pathways. Neuroinflammation is closely associated with the pathogenesis of neurodegenerative diseases, neurogenic tumors and cerebrovascular diseases. From recent studies, it is clear that FPRs are important biomarkers for neurological diseases as they regulate inflammatory responses by monitoring glial activation, accelerating neural differentiation, regulating angiogenesis, and controlling blood brain barrier (BBB) permeability, thereby affecting neurological disease progression. Given the complex mechanisms of neurological diseases and the difficulty of healing, we are eager to find new and effective therapeutic targets. Here, we review recent research about various mechanisms of the effects generated after FPR binding to different ligands, role of FPRs in neuroinflammation as well as the development and prognosis of neurological diseases. We summarize that the FPR family has dual inflammatory functional properties in central nervous system. Emphasizing that FPR2 acts as a key molecule that mediates the active resolution of inflammation, which binds with corresponding receptors to reduce the expression and activation of pro-inflammatory composition, govern the transport of immune cells to inflammatory tissues, and restore the integrity of the BBB. Concurrently, FPR1 is essentially related to angiogenesis, cell proliferation and neurogenesis. Thus, treatment with FPRs-modulation may be effective for neurological diseases.
Collapse
Affiliation(s)
- Jiahui Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingfei Li
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiao Ding
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinyu Huang
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Tang
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Busch L, Vieten S, Brödel S, Endres K, Bufe B. Emerging contributions of formyl peptide receptors to neurodegenerative diseases. Biol Chem 2021; 403:27-41. [PMID: 34505459 DOI: 10.1515/hsz-2021-0258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/23/2021] [Indexed: 12/28/2022]
Abstract
Inflammation is a central element of many neurodegenerative diseases. Formyl peptide receptors (FPRs) can trigger several receptor-dependent signal transduction pathways that play a key role in neuroinflammation and neurodegeneration. They are chemotactic receptors that help to regulate pro- and anti-inflammatory responses in most mammals. FPRs are primarily expressed in the immune and nervous systems where they interact with a complex pattern of pathogen-derived and host-endogenous molecules. Mounting evidence points towards a contribution of FPRs - via neuropathological ligands such as Amyloid beta, and neuroprotective ligands such as Humanin, Lipoxin A4, and Annexin A1 - to multiple pathological aspects of neurodegenerative diseases. In this review, we aim to summarize the interplay of FPRs with neuropathological and neuroprotective ligands. Next, we depict their capability to trigger a number of ligand-dependent cell signaling pathways and their potential to interact with additional intracellular cofactors. Moreover, we highlight first studies, demonstrating that a pharmacological inhibition of FPRs helps to ameliorate neuroinflammation, which may pave the way towards novel therapeutic strategies.
Collapse
Affiliation(s)
- Lukas Busch
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Stefan Vieten
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Susan Brödel
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Centre of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Bernd Bufe
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
4
|
Formyl peptide receptor 2, as an important target for ligands triggering the inflammatory response regulation: a link to brain pathology. Pharmacol Rep 2021; 73:1004-1019. [PMID: 34105114 PMCID: PMC8413167 DOI: 10.1007/s43440-021-00271-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/13/2021] [Accepted: 04/30/2021] [Indexed: 12/28/2022]
Abstract
Formyl peptide receptors (FPRs) belong to the family of seven-transmembrane G protein-coupled receptors. Among them, FPR2 is a low affinity receptor for N-formyl peptides and is considered the most promiscuous member of FPRs. FPR2 is able to recognize a broad variety of endogenous or exogenous ligands, ranging from lipid to proteins and peptides, including non-formylated peptides. Due to this property FPR2 has the ability to modulate both pro- and anti-inflammatory response, depending on the nature of the bound agonist and on the different recognition sites of the receptor. Thus, FPR2 takes part not only in the proinflammatory response but also in the resolution of inflammation (RoI) processes. Recent data have indicated that the malfunction of RoI may be the background for some central nervous system (CNS) disorders. Therefore, much interest is focused on endogenous molecules called specialized pro-resolving mediators (SPMs), as well as on new synthetic FPR2 agonists, which kick-start the resolution of inflammation (RoI) and modulate its course. Here, we shed some light on the general characteristics of the FPR family in humans and in the experimental animals. Moreover, we present a guide to understanding the "double faced" action of FPR2 activation in the context of immune-related diseases of the CNS.
Collapse
|
5
|
Chemotactic Ligands that Activate G-Protein-Coupled Formylpeptide Receptors. Int J Mol Sci 2019; 20:ijms20143426. [PMID: 31336833 PMCID: PMC6678346 DOI: 10.3390/ijms20143426] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 12/14/2022] Open
Abstract
Leukocyte infiltration is a hallmark of inflammatory responses. This process depends on the bacterial and host tissue-derived chemotactic factors interacting with G-protein-coupled seven-transmembrane receptors (GPCRs) expressed on the cell surface. Formylpeptide receptors (FPRs in human and Fprs in mice) belong to the family of chemoattractant GPCRs that are critical mediators of myeloid cell trafficking in microbial infection, inflammation, immune responses and cancer progression. Both murine Fprs and human FPRs participate in many patho-physiological processes due to their expression on a variety of cell types in addition to myeloid cells. FPR contribution to numerous pathologies is in part due to its capacity to interact with a plethora of structurally diverse chemotactic ligands. One of the murine Fpr members, Fpr2, and its endogenous agonist peptide, Cathelicidin-related antimicrobial peptide (CRAMP), control normal mouse colon epithelial growth, repair and protection against inflammation-associated tumorigenesis. Recent developments in FPR (Fpr) and ligand studies have greatly expanded the scope of these receptors and ligands in host homeostasis and disease conditions, therefore helping to establish these molecules as potential targets for therapeutic intervention.
Collapse
|
6
|
Raabe CA, Gröper J, Rescher U. Biased perspectives on formyl peptide receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:305-316. [DOI: 10.1016/j.bbamcr.2018.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023]
|
7
|
Buchan KD, Foster SJ, Renshaw SA. Staphylococcus aureus: setting its sights on the human innate immune system. MICROBIOLOGY-SGM 2019; 165:367-385. [PMID: 30625113 DOI: 10.1099/mic.0.000759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Staphylococcus aureus has colonized humans for at least 10 000 years, and today inhabits roughly a third of the population. In addition, S. aureus is a major pathogen that is responsible for a significant disease burden, ranging in severity from mild skin and soft-tissue infections to life-threatening endocarditis and necrotizing pneumonia, with treatment often hampered by resistance to commonly available antibiotics. Underpinning its versatility as a pathogen is its ability to evade the innate immune system. S. aureus specifically targets innate immunity to establish and sustain infection, utilizing a large repertoire of virulence factors to do so. Using these factors, S. aureus can resist phagosomal killing, impair complement activity, disrupt cytokine signalling and target phagocytes directly using proteolytic enzymes and cytolytic toxins. Although most of these virulence factors are well characterized, their importance during infection is less clear, as many display species-specific activity against humans or against animal hosts, including cows, horses and chickens. Several staphylococcal virulence factors display species specificity for components of the human innate immune system, with as few as two amino acid changes reducing binding affinity by as much as 100-fold. This represents a major issue for studying their roles during infection, which cannot be examined without the use of humanized infection models. This review summarizes the major factors S. aureus uses to impair the innate immune system, and provides an in-depth look into the host specificity of S. aureus and how this problem is being approached.
Collapse
Affiliation(s)
- Kyle D Buchan
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Simon J Foster
- 2Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Stephen A Renshaw
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
8
|
Ilitchev AI, Giammona MJ, Olivas C, Claud SL, Lazar Cantrell KL, Wu C, Buratto SK, Bowers MT. Hetero-oligomeric Amyloid Assembly and Mechanism: Prion Fragment PrP(106-126) Catalyzes the Islet Amyloid Polypeptide β-Hairpin. J Am Chem Soc 2018; 140:9685-9695. [PMID: 29989407 DOI: 10.1021/jacs.8b05925] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein aggregation is typically attributed to the association of homologous amino acid sequences between monomers of the same protein. Coaggregation of heterogeneous peptide species can occur, however, and is implicated in the proliferation of seemingly unrelated protein diseases in the body. The prion protein fragment (PrP106-126) and human islet amyloid polypeptide (hIAPP) serve as an interesting model of nonhomologous protein assembly as they coaggregate, despite a lack of sequence homology. We have applied ion-mobility mass spectrometry, atomic force microscopy, circular dichroism, and high-level molecular modeling to elucidate this important assembly process. We found that the prion fragment not only forms pervasive hetero-oligomeric aggregates with hIAPP but also promotes the transition of hIAPP into its amyloidogenic β-hairpin conformation. Further, when PrP106-126 was combined with non-amyloidogenic rIAPP, the two formed nearly identical hetero-oligomers to those seen with hIAPP, despite rIAPP containing β-sheet breaking proline substitutions. Additionally, while rIAPP does not natively form the amyloidogenic β-hairpin structure, it did so in the presence of PrP106-126 and underwent a conformational transition to β-sheet in solution. We also find that PrP106-126 forms hetero-oligomers with the IAPP8-20 fragment but not with the "aggregation hot spot" IAPP20-29 fragment. PrP106-126 apparently induces IAPP into a β-hairpin structure within the PrP:IAPP heterodimer complex and then, through ligand exchange, catalytically creates the amyloidogenic β-hairpin dimer of IAPP in significantly greater abundance than IAPP does on its own. This is a new mechanistic model that provides a critical foundation for the detailed study of hetero-oligomerization and prion-like proliferation in amyloid systems.
Collapse
Affiliation(s)
- Alexandre I Ilitchev
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , United States
| | - Maxwell J Giammona
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , United States
| | - Carina Olivas
- Department of Chemistry and Biochemistry , Rowan University , Glassboro , New Jersey 08028 , United States
| | - Sarah L Claud
- Department of Chemistry , Westmont College , Santa Barbara , California 93108 , United States
| | - Kristi L Lazar Cantrell
- Department of Chemistry , Westmont College , Santa Barbara , California 93108 , United States
| | - Chun Wu
- Department of Chemistry and Biochemistry , Rowan University , Glassboro , New Jersey 08028 , United States
| | - Steven K Buratto
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , United States
| | - Michael T Bowers
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , United States
| |
Collapse
|
9
|
Targeting formyl peptide receptors to facilitate the resolution of inflammation. Eur J Pharmacol 2018; 833:339-348. [PMID: 29935171 DOI: 10.1016/j.ejphar.2018.06.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
The formyl peptide receptors (FPRs) are G protein coupled receptors that recognize a broad range of structurally distinct pathogen and danger-associated molecular patterns and mediate host defense to infection and tissue injury. It became evident that the cellular distribution and biological functions of FPRs extend beyond myeloid cells and governing their activation and trafficking. In recent years, significant progress has been made to position FPRs at check points that control the resolution of inflammation, tissue repair and return to homeostasis. Accumulating data indicate a role for FPRs in an ever-increasing range of human diseases, including atherosclerosis, chronic obstructive pulmonary disease, asthma, autoimmune diseases and cancer, in which dysregulated or defective resolution are increasingly recognized as critical component of the pathogenesis. This review summarizes recent advances on how FPRs recognize distinct ligands and integrate opposing cues to govern various responses and will discuss how this knowledge could be harnessed for developing novel therapeutic strategies to counter inflammation that underlies many human diseases.
Collapse
|
10
|
fMLP-dependent activation of Akt and ERK1/2 through ROS/Rho A pathways is mediated through restricted activation of the FPRL1 (FPR2) receptor. Inflamm Res 2018; 67:711-722. [PMID: 29922854 DOI: 10.1007/s00011-018-1163-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 05/02/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE AND DESIGN The objective of this study is to uncover the signal transduction pathways of N-formyl methionyl-leucyl-phenylalanine (fMLP) in monocyte. MATERIALS OR SUBJECTS Freshly isolated human peripheral blood monocytes (PBMC) were used for in vitro assessment of signal transduction pathways activated by fMLP. TREATMENT Time-course and dose-response experiments were used to evaluate the effect of fMLP along with the specific inhibitors/stimulators on the activation of downstream signaling kinases. METHODS Freshly isolated human PBMC were stimulated with fMLP for the desired time. Western blot and siRNA analysis were used to evaluate the activated intracellular signaling kinases, and flow analysis was performed to assess the levels of CD11b. Furthermore, luminescence spectrometry was performed to measure the levels of released hydrogen peroxide in the media. RESULTS fMLP strongly stimulated the activation of AKT and ERK1/2 through a RhoA-GTPase-dependent manner and also induced H2O2 release by monocytes. Furthermore, fMLP mediated its effects through restricted activation of formylpeptide receptor-like 1 (FPRL1/FPR2), but independently of either EGFR transactivation or intracellular calcium release. In addition, NAC reversed fMLP- and H2O2-induced activation of Akt and RhoA-GTPase. CONCLUSION Collectively, these data suggested that fMLP-activated ERK1/2 and Akt pathways through specific activation of the FPRL1/ROS/RoA-GTPase pathway.
Collapse
|
11
|
Guerra FE, Borgogna TR, Patel DM, Sward EW, Voyich JM. Epic Immune Battles of History: Neutrophils vs. Staphylococcus aureus. Front Cell Infect Microbiol 2017; 7:286. [PMID: 28713774 PMCID: PMC5491559 DOI: 10.3389/fcimb.2017.00286] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/12/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and the first line of defense after bacteria have breached the epithelial barriers. After migration to a site of infection, neutrophils engage and expose invading microorganisms to antimicrobial peptides and proteins, as well as reactive oxygen species, as part of their bactericidal arsenal. Ideally, neutrophils ingest bacteria to prevent damage to surrounding cells and tissues, kill invading microorganisms with antimicrobial mechanisms, undergo programmed cell death to minimize inflammation, and are cleared away by macrophages. Staphylococcus aureus (S. aureus) is a prevalent Gram-positive bacterium that is a common commensal and causes a wide range of diseases from skin infections to endocarditis. Since its discovery, S. aureus has been a formidable neutrophil foe that has challenged the efficacy of this professional assassin. Indeed, proper clearance of S. aureus by neutrophils is essential to positive infection outcome, and S. aureus has developed mechanisms to evade neutrophil killing. Herein, we will review mechanisms used by S. aureus to modulate and evade neutrophil bactericidal mechanisms including priming, activation, chemotaxis, production of reactive oxygen species, and resolution of infection. We will also highlight how S. aureus uses sensory/regulatory systems to tailor production of virulence factors specifically to the triggering signal, e.g., neutrophils and defensins. To conclude, we will provide an overview of therapeutic approaches that may potentially enhance neutrophil antimicrobial functions.
Collapse
Affiliation(s)
- Fermin E Guerra
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Timothy R Borgogna
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Delisha M Patel
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Eli W Sward
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State UniversityBozeman, MT, United States
| |
Collapse
|
12
|
Hou XL, Ji CD, Tang J, Wang YX, Xiang DF, Li HQ, Liu WW, Wang JX, Yan HZ, Wang Y, Zhang P, Cui YH, Wang JM, Bian XW, Liu W. FPR2 promotes invasion and metastasis of gastric cancer cells and predicts the prognosis of patients. Sci Rep 2017; 7:3153. [PMID: 28600569 PMCID: PMC5466646 DOI: 10.1038/s41598-017-03368-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/27/2017] [Indexed: 01/20/2023] Open
Abstract
Formyl peptide receptor 2 (FPR2), a classical chemoattractant receptor of G-protein-coupled receptors, is reported to be involved in invasion and metastasis of some cancers, but the role of FPR2 in gastric cancer (GC) has not yet been elucidated. In this study, we found that the levels of FPR2 expression in GC were positively correlated with invasion depth, lymph node metastasis and negatively correlated with the patients’ overall survival. Multivariate analysis indicated that FPR2 expression was an independent prognostic marker for GC patients. FPR2-knockdown significantly abrogated the migration and invasion stimulated by Hp(2–20) and Ac(2–26), two well-characterized ligands for FPR2 in GC cells. FPR2 deletion also reduced the tumorigenic and metastatic capabilities of GC cells in vivo. Mechanistically, stimulation with FPR2 ligands resulted in down-regulation of E-cadherin and up-regulation of vimentin, which were reversed by FPR2 knock-down, implying the involvement of epithelial–mesenchymal transition (EMT). Moreover, the activation of FPR2 was accompanied with ERK1/2 phosphorylation, which could be attenuated by FPR2 silencing or treatment with MEK inhibitor, PD98059. Altogether, our results demonstrate that FPR2 is functionally involved in invasion and metastasis, and potentially acts as a novel prognostic marker as well as a potential therapeutic target in human GC.
Collapse
Affiliation(s)
- Xi-Lu Hou
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - Cheng-Dong Ji
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - Jun Tang
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - Yan-Xia Wang
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - Dong-Fang Xiang
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - Hai-Qing Li
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - Wei-Wei Liu
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - Jiao-Xue Wang
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - He-Zhong Yan
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China
| | - Yan Wang
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - Peng Zhang
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - You-Hong Cui
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China
| | - Ji-Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Xiu-Wu Bian
- Institute of Pathology & Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Third Military Medical University, Chongqing, 400038, China.
| | - Wei Liu
- Department of Gastroenterology, The 105th Hospital of People's Liberation Army, Hefei, Anhui, 230031, China.
| |
Collapse
|
13
|
He HQ, Ye RD. The Formyl Peptide Receptors: Diversity of Ligands and Mechanism for Recognition. Molecules 2017; 22:E455. [PMID: 28335409 PMCID: PMC6155412 DOI: 10.3390/molecules22030455] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 03/09/2017] [Indexed: 12/21/2022] Open
Abstract
The formyl peptide receptors (FPRs) are G protein-coupled receptors that transduce chemotactic signals in phagocytes and mediate host-defense as well as inflammatory responses including cell adhesion, directed migration, granule release and superoxide production. In recent years, the cellular distribution and biological functions of FPRs have expanded to include additional roles in homeostasis of organ functions and modulation of inflammation. In a prototype, FPRs recognize peptides containing N-formylated methionine such as those produced in bacteria and mitochondria, thereby serving as pattern recognition receptors. The repertoire of FPR ligands, however, has expanded rapidly to include not only N-formyl peptides from microbes but also non-formyl peptides of microbial and host origins, synthetic small molecules and an eicosanoid. How these chemically diverse ligands are recognized by the three human FPRs (FPR1, FPR2 and FPR3) and their murine equivalents is largely unclear. In the absence of crystal structures for the FPRs, site-directed mutagenesis, computer-aided ligand docking and structural simulation have led to the identification of amino acids within FPR1 and FPR2 that interact with several formyl peptides. This review article summarizes the progress made in the understanding of FPR ligand diversity as well as ligand recognition mechanisms used by these receptors.
Collapse
Affiliation(s)
- Hui-Qiong He
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
- Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| | - Richard D Ye
- Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
14
|
Tu J, Chen B, Yang L, Qi K, Lu J, Zhao D. Amyloid-β Activates Microglia and Regulates Protein Expression in a Manner Similar to Prions. J Mol Neurosci 2015; 56:509-18. [PMID: 25869610 DOI: 10.1007/s12031-015-0553-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
Prions are the only convincingly demonstrated proteinaceous infectious particle, yet recent studies find that amyloid-β peptide (Aβ) aggregates are capable of self-propagation, which induces amyloidosis pathology in Alzheimer's disease (AD) model mice that is similar to the self-propagation phenomenon of prions in neurons. Gliosis is a common hallmark of AD and prion diseases, in which activated microglia accumulate around abnormal protein deposits. Analyses of the characteristics of activated microglia induced by Aβ in comparison with those induced by prions will provide new insight into the pathogenesis of AD. Therefore, we compared the characteristics of BV-2 cells (model microglia) activated by Aβ fibrillar peptides (Aβ1-42) and prions (PrP106-126). Aβ1-42 and PrP106-126, as well as the supernatants of the media collected from BV-2 cells cocultured with Aβ1-42 and PrP106-126, were potent activators of BV-2 microglial activity, but the chemotaxis index (CI) induced by Aβ1-42 was significantly higher than that induced by PrP106-126 at each concentration. Aβ1-42 and PrP106-126 increased the proliferation index (PI) and upregulated monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor beta 1 (TGF-β1) expression after 12 h of exposure. Our results show that Aβ activates microglia and regulates microglial protein expression in a manner similar to prions and, thus, provide new insight into the pathogenesis of AD.
Collapse
Affiliation(s)
- Jian Tu
- State Key Laboratories for Agrobiotechnology, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China,
| | | | | | | | | | | |
Collapse
|
15
|
Cash JL, Norling LV, Perretti M. Resolution of inflammation: targeting GPCRs that interact with lipids and peptides. Drug Discov Today 2014; 19:1186-92. [PMID: 24993159 PMCID: PMC4154450 DOI: 10.1016/j.drudis.2014.06.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/23/2014] [Indexed: 11/30/2022]
Abstract
There is a growing appreciation of the important role of resolution mediators in the successful termination of the inflammatory response. Here, we discuss the potential importance of the lipid and peptide proresolving mediators, in particular the resolvins and chemerin-derived peptides, which mediate their effects through specific G protein-coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Jenna L Cash
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, UK
| | - Lucy V Norling
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, UK
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
16
|
Song K, Na JY, Oh MH, Kim S, Kim YH, Park BY, Shin GW, Kim B, You M, Kwon J. Synthetic prion Peptide 106-126 resulted in an increase matrix metalloproteinases and inflammatory cytokines from rat astrocytes and microglial cells. Toxicol Res 2013; 28:5-9. [PMID: 24278583 PMCID: PMC3834397 DOI: 10.5487/tr.2012.28.1.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/12/2012] [Accepted: 03/21/2012] [Indexed: 12/22/2022] Open
Abstract
It has been shown that the accumulation of prion in the cytoplasm can result in neurodegenerative disorders. Synthetic prion peptide 106-126 (PrP) is a glycoprotein that is expressed predominantly by neurons and other cells, including glial cells. Prion-induced chronic neurodegeneration has a substantial inflammatory component, and an increase in the levels of matrix metalloproteinases (MMPs) may play an important role in neurodegenerative development and progression. However, the expression of MMPs in PrP induced rat astrocytes and microglia has not yet been compared. Thus, in this study, we examined the fluorescence intensity of CD11b positive microglia and Glial Fibrillary Acidic Protein (GFAP) positive astrocytes and found that the fluorescent intensity was increased following incubation with PrP at 24 hours in a dose-dependent manner. We also observed an increase in interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) protein expression, which are initial inflammatory cytokines, in both PrP induced astrocytes and microglia. Furthermore, an increase MMP-1, 3 and 11 expressions in PrP induced astrocytes and microglia was observed by real time PCR. Our results demonstrated PrP induced activation of astrocytes and microglia respectively, which resulted in an increase in inflammatory cytokines and MMPs expression. These results provide the insight into the different sensitivities of glial cells to PrP.
Collapse
Affiliation(s)
- Kibbeum Song
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-156, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Tu J, Yang L, Zhou X, Qi K, Wang J, Kouadir M, Xu L, Yin X, Zhao D. PrP106-126 and Aβ1-42 Peptides Induce BV-2 Microglia Chemotaxis and Proliferation. J Mol Neurosci 2013; 52:107-16. [DOI: 10.1007/s12031-013-0140-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/04/2013] [Indexed: 11/30/2022]
|
18
|
Vilches S, Vergara C, Nicolás O, Sanclimens G, Merino S, Varón S, Acosta GA, Albericio F, Royo M, Río JAD, Gavín R. Neurotoxicity of prion peptides mimicking the central domain of the cellular prion protein. PLoS One 2013; 8:e70881. [PMID: 23940658 PMCID: PMC3733940 DOI: 10.1371/journal.pone.0070881] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/25/2013] [Indexed: 12/20/2022] Open
Abstract
The physiological functions of PrP(C) remain enigmatic, but the central domain, comprising highly conserved regions of the protein may play an important role. Indeed, a large number of studies indicate that synthetic peptides containing residues 106-126 (CR) located in the central domain (CD, 95-133) of PrP(C) are neurotoxic. The central domain comprises two chemically distinct subdomains, the charge cluster (CC, 95-110) and a hydrophobic region (HR, 112-133). The aim of the present study was to establish the individual cytotoxicity of CC, HR and CD. Our results show that only the CD peptide is neurotoxic. Biochemical, Transmission Electron Microscopy and Atomic Force Microscopy experiments demonstrated that the CD peptide is able to activate caspase-3 and disrupt the cell membrane, leading to cell death.
Collapse
Affiliation(s)
- Silvia Vilches
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Cristina Vergara
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Oriol Nicolás
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Gloria Sanclimens
- Combinatorial Chemistry Unit, Scientific Park of Barcelona, Barcelona, Spain
| | - Sandra Merino
- Department of Physicochemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Sonia Varón
- Combinatorial Chemistry Unit, Scientific Park of Barcelona, Barcelona, Spain
| | - Gerardo A. Acosta
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Fernando Albericio
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
- Department of Organic Chemistry, Faculty of Chemistry, University of Barcelona, Barcelona, Spain
| | - Miriam Royo
- Combinatorial Chemistry Unit, Scientific Park of Barcelona, Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - José A. Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Pinilla C, Edwards BS, Appel JR, Yates-Gibbins T, Giulianotti MA, Medina-Franco JL, Young SM, Santos RG, Sklar LA, Houghten RA. Selective agonists and antagonists of formylpeptide receptors: duplex flow cytometry and mixture-based positional scanning libraries. Mol Pharmacol 2013; 84:314-24. [PMID: 23788657 DOI: 10.1124/mol.113.086595] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The formylpeptide receptor (FPR1) and formylpeptide-like 1 receptor (FPR2) are G protein-coupled receptors that are linked to acute inflammatory responses, malignant glioma stem cell metastasis, and chronic inflammation. Although several N-formyl peptides are known to bind to these receptors, more selective small-molecule, high-affinity ligands are needed for a better understanding of the physiologic roles played by these receptors. High-throughput assays using mixture-based combinatorial libraries represent a unique, highly efficient approach for rapid data acquisition and ligand identification. We report the superiority of this approach in the context of the simultaneous screening of a diverse set of mixture-based small-molecule libraries. We used a single cross-reactive peptide ligand for a duplex flow cytometric screen of FPR1 and FPR2 in color-coded cell lines. Screening 37 different mixture-based combinatorial libraries totaling more than five million small molecules (contained in 5,261 mixture samples) resulted in seven libraries that significantly inhibited activity at the receptors. Using positional scanning deconvolution, selective high-affinity (low nM K(i)) individual compounds were identified from two separate libraries, namely, pyrrolidine bis-diketopiperazine and polyphenyl urea. The most active individual compounds were characterized for their functional activities as agonists or antagonists with the most potent FPR1 agonist and FPR2 antagonist identified to date with an EC₅₀ of 131 nM (4 nM K(i)) and an IC₅₀ of 81 nM (1 nM K(i)), respectively, in intracellular Ca²⁺ response determinations. Comparative analyses of other previous screening approaches clearly illustrate the efficiency of identifying receptor selective, individual compounds from mixture-based combinatorial libraries.
Collapse
Affiliation(s)
- Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, San Diego, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Medina-Franco JL, Edwards BS, Pinilla C, Appel JR, Giulianotti MA, Santos RG, Yongye AB, Sklar LA, Houghten RA. Rapid scanning structure-activity relationships in combinatorial data sets: identification of activity switches. J Chem Inf Model 2013; 53:1475-85. [PMID: 23705689 DOI: 10.1021/ci400192y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We present a general approach to describe the structure-activity relationships (SAR) of combinatorial data sets with activity for two biological endpoints with emphasis on the rapid identification of substitutions that have a large impact on activity and selectivity. The approach uses dual-activity difference (DAD) maps that represent a visual and quantitative analysis of all pairwise comparisons of one, two, or more substitutions around a molecular template. Scanning the SAR of data sets using DAD maps allows the visual and quantitative identification of activity switches defined as specific substitutions that have an opposite effect on the activity of the compounds against two targets. The approach also rapidly identifies single- and double-target R-cliffs, i.e., compounds where a single or double substitution around the central scaffold dramatically modifies the activity for one or two targets, respectively. The approach introduced in this report can be applied to any analogue series with two biological activity endpoints. To illustrate the approach, we discuss the SAR of 106 pyrrolidine bis-diketopiperazines tested against two formylpeptide receptors obtained from positional scanning deconvolution methods of mixture-based libraries.
Collapse
Affiliation(s)
- José L Medina-Franco
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida 34987, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The mathematics of a successful deconvolution: a quantitative assessment of mixture-based combinatorial libraries screened against two formylpeptide receptors. Molecules 2013; 18:6408-24. [PMID: 23722730 PMCID: PMC4106117 DOI: 10.3390/molecules18066408] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 01/04/2023] Open
Abstract
In the past 20 years, synthetic combinatorial methods have fundamentally advanced the ability to synthesize and screen large numbers of compounds for drug discovery and basic research. Mixture-based libraries and positional scanning deconvolution combine two approaches for the rapid identification of specific scaffolds and active ligands. Here we present a quantitative assessment of the screening of 32 positional scanning libraries in the identification of highly specific and selective ligands for two formylpeptide receptors. We also compare and contrast two mixture-based library approaches using a mathematical model to facilitate the selection of active scaffolds and libraries to be pursued for further evaluation. The flexibility demonstrated in the differently formatted mixture-based libraries allows for their screening in a wide range of assays.
Collapse
|
22
|
Distinct signaling cascades elicited by different formyl peptide receptor 2 (FPR2) agonists. Int J Mol Sci 2013; 14:7193-230. [PMID: 23549262 PMCID: PMC3645683 DOI: 10.3390/ijms14047193] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/13/2013] [Accepted: 03/15/2013] [Indexed: 12/22/2022] Open
Abstract
The formyl peptide receptor 2 (FPR2) is a remarkably versatile transmembrane protein belonging to the G-protein coupled receptor (GPCR) family. FPR2 is activated by an array of ligands, which include structurally unrelated lipids and peptide/proteins agonists, resulting in different intracellular responses in a ligand-specific fashion. In addition to the anti-inflammatory lipid, lipoxin A4, several other endogenous agonists also bind FPR2, including serum amyloid A, glucocorticoid-induced annexin 1, urokinase and its receptor, suggesting that the activation of FPR2 may result in potent pro- or anti-inflammatory responses. Other endogenous ligands, also present in biological samples, include resolvins, amyloidogenic proteins, such as beta amyloid (Aβ)-42 and prion protein (Prp)106–126, the neuroprotective peptide, humanin, antibacterial peptides, annexin 1-derived peptides, chemokine variants, the neuropeptides, vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP)-27, and mitochondrial peptides. Upon activation, intracellular domains of FPR2 mediate signaling to G-proteins, which trigger several agonist-dependent signal transduction pathways, including activation of phospholipase C (PLC), protein kinase C (PKC) isoforms, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, the mitogen-activated protein kinase (MAPK) pathway, p38MAPK, as well as the phosphorylation of cytosolic tyrosine kinases, tyrosine kinase receptor transactivation, phosphorylation and nuclear translocation of regulatory transcriptional factors, release of calcium and production of oxidants. FPR2 is an attractive therapeutic target, because of its involvement in a range of normal physiological processes and pathological diseases. Here, we review and discuss the most significant findings on the intracellular pathways and on the cross-communication between FPR2 and tyrosine kinase receptors triggered by different FPR2 agonists.
Collapse
|
23
|
Brennan EP, Nolan KA, Börgeson E, Gough OS, McEvoy CM, Docherty NG, Higgins DF, Murphy M, Sadlier DM, Ali-Shah ST, Guiry PJ, Savage DA, Maxwell AP, Martin F, Godson C. Lipoxins attenuate renal fibrosis by inducing let-7c and suppressing TGFβR1. J Am Soc Nephrol 2013; 24:627-37. [PMID: 23520204 DOI: 10.1681/asn.2012060550] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lipoxins, which are endogenously produced lipid mediators, promote the resolution of inflammation, and may inhibit fibrosis, suggesting a possible role in modulating renal disease. Here, lipoxin A4 (LXA4) attenuated TGF-β1-induced expression of fibronectin, N-cadherin, thrombospondin, and the notch ligand jagged-1 in cultured human proximal tubular epithelial (HK-2) cells through a mechanism involving upregulation of the microRNA let-7c. Conversely, TGF-β1 suppressed expression of let-7c. In cells pretreated with LXA4, upregulation of let-7c persisted despite subsequent stimulation with TGF-β1. In the unilateral ureteral obstruction model of renal fibrosis, let-7c upregulation was induced by administering an LXA4 analog. Bioinformatic analysis suggested that targets of let-7c include several members of the TGF-β1 signaling pathway, including the TGF-β receptor type 1. Consistent with this, LXA4-induced upregulation of let-7c inhibited both the expression of TGF-β receptor type 1 and the response to TGF-β1. Overexpression of let-7c mimicked the antifibrotic effects of LXA4 in renal epithelia; conversely, anti-miR directed against let-7c attenuated the effects of LXA4. Finally, we observed that several let-7c target genes were upregulated in fibrotic human renal biopsies compared with controls. In conclusion, these results suggest that LXA4-mediated upregulation of let-7c suppresses TGF-β1-induced fibrosis and that expression of let-7c targets is dysregulated in human renal fibrosis.
Collapse
Affiliation(s)
- Eoin P Brennan
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Edwards BS, Young SM, Saunders MJ, Bologa C, Oprea TI, Ye RD, Prossnitz ER, Graves SW, Sklar LA. High-throughput flow cytometry for drug discovery. Expert Opin Drug Discov 2013; 2:685-96. [PMID: 23488958 DOI: 10.1517/17460441.2.5.685] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
High-throughput flow cytometry exploits a novel many-samples/one-file approach to dramatically speed data acquisition, limit aspirated sample volume to as little as 2 μl/well and produce multisample data sets that facilitate automated analysis of samples in groups as well as individually. It has been successfully applied to both cell- and microsphere-based bioassays in 96- and 384-well formats, to screen tens-of-thousands of compounds and identify novel bioactive structures. High-content multiparametric analysis capabilities have been exploited for assay multiplexing, allowing the assessment of biologic selectivity and specificity to be an integral component of primary screens. These and other advances in the last decade have contributed to the application of flow cytometry as a uniquely powerful tool for probing biologic and chemical diversity and complex systems biology.
Collapse
Affiliation(s)
- Bruce S Edwards
- Cytometry and Department of Pathology, CRTC, UNM HSC, MS08-4630, Albuquerque, NM 87131, USA +1 505 272 6206 ; +1 505 272 6695 ;
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Li Y, Ye D. Molecular biology for formyl peptide receptors in human diseases. J Mol Med (Berl) 2013; 91:781-9. [PMID: 23404331 DOI: 10.1007/s00109-013-1005-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 01/21/2013] [Accepted: 01/27/2013] [Indexed: 02/07/2023]
Abstract
Leukocytes accumulate at sites of inflammation and immunological reaction in response to locally existing chemotactic mediators. The first chemotactic factors structurally defined were N-formyl peptides. Subsequently, numerous ligands were identified to activate formyl peptide receptors (FPRs) that belong to the seven-transmembrane G protein-coupled receptor superfamily. FPRs interact with this menagerie of structurally diverse pro- and anti-inflammatory ligands to possess important regulatory effects in multiple diseases, including inflammation, amyloidosis, Alzheimer's disease, prion disease, acquired immunodeficiency syndrome, obesity, diabetes, and cancer. How these receptors recognize diverse ligands and how they contribute to disease pathogenesis and host defense are basic questions currently under investigation that would open up new avenues for the future management of inflammation-related diseases.
Collapse
Affiliation(s)
- Yongsheng Li
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China.
| | | |
Collapse
|
26
|
Bradford BM, Mabbott NA. Prion disease and the innate immune system. Viruses 2012; 4:3389-419. [PMID: 23342365 PMCID: PMC3528271 DOI: 10.3390/v4123389] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 11/14/2012] [Accepted: 11/22/2012] [Indexed: 02/06/2023] Open
Abstract
Prion diseases or transmissible spongiform encephalopathies are a unique category of infectious protein-misfolding neurodegenerative disorders. Hypothesized to be caused by misfolding of the cellular prion protein these disorders possess an infectious quality that thrives in immune-competent hosts. While much has been discovered about the routing and critical components involved in the peripheral pathogenesis of these agents there are still many aspects to be discovered. Research into this area has been extensive as it represents a major target for therapeutic intervention within this group of diseases. The main focus of pathological damage in these diseases occurs within the central nervous system. Cells of the innate immune system have been proven to be critical players in the initial pathogenesis of prion disease, and may have a role in the pathological progression of disease. Understanding how prions interact with the host innate immune system may provide us with natural pathways and mechanisms to combat these diseases prior to their neuroinvasive stage. We present here a review of the current knowledge regarding the role of the innate immune system in prion pathogenesis.
Collapse
Affiliation(s)
- Barry M Bradford
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | | |
Collapse
|
27
|
Incunabular immunological events in prion trafficking. Sci Rep 2012; 2:440. [PMID: 22679554 PMCID: PMC3368226 DOI: 10.1038/srep00440] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/21/2012] [Indexed: 11/08/2022] Open
Abstract
While prions probably interact with the innate immune system immediately following infection, little is known about this initial confrontation. Here we investigated incunabular events in lymphotropic and intranodal prion trafficking by following highly enriched, fluorescent prions from infection sites to draining lymph nodes. We detected biphasic lymphotropic transport of prions from the initial entry site upon peripheral prion inoculation. Prions arrived in draining lymph nodes cell autonomously within two hours of intraperitoneal administration. Monocytes and dendritic cells (DCs) required Complement for optimal prion delivery to lymph nodes hours later in a second wave of prion trafficking. B cells constituted the majority of prion-bearing cells in the mediastinal lymph node by six hours, indicating intranodal prion reception from resident DCs or subcapsulary sinus macrophages or directly from follicular conduits. These data reveal novel, cell autonomous prion lymphotropism, and a prominent role for B cells in intranodal prion movement.
Collapse
|
28
|
Prion Peptide PrP106-126 Induces Inducible Nitric Oxide Synthase and Proinflammatory Cytokine Gene Expression Through the Activation of NF-κB in Macrophage Cells. DNA Cell Biol 2012; 31:833-8. [DOI: 10.1089/dna.2011.1362] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
29
|
Yuan L, Zhou X, Li D, Ma W, Yu H, Xi Y, Xiao R. Pattern recognition receptors involved in the inflammatory attenuating effects of soybean isoflavone in β-amyloid peptides 1-42 treated rats. Neurosci Lett 2012; 506:266-70. [PMID: 22133809 DOI: 10.1016/j.neulet.2011.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 10/14/2011] [Accepted: 11/13/2011] [Indexed: 11/19/2022]
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 β-amyloid peptides1-42 (Aβ1-42). In present study, we further detected the effects of SIF against inflammation caused by Aβ1-42 treatment in rats. Serum inflammatory mediators and neurotrophic factors including transforming growth factor-β (TGF-β), inducible nitric oxide synthase (iNOS), brain-derived neurotrophic factor (BDNF) and S100β 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-1β (IL-1β), iNOS, tumor necrosis factor-α (TNF-α), TGF-β, BDNF, S100β, 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-1β, TNF-α and iNOS induced by Aβ1-42 in serum and brain of rats. SIF also significantly reversed Aβ1-42-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 Aβ1-42 treatment, and SIF was an efficiency compound in reversing the inflammation caused by Aβ1-42 treatment.
Collapse
Affiliation(s)
- Linhong Yuan
- Department of Nutrition and Food Hygiene, Capital Medical University, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
30
|
Stranahan AM, Martin B, Chadwick W, Park SS, Wang L, Becker KG, WoodIII WH, Zhang Y, Maudsley S. Metabolic context regulates distinct hypothalamic transcriptional responses to antiaging interventions. Int J Endocrinol 2012; 2012:732975. [PMID: 22934110 PMCID: PMC3427989 DOI: 10.1155/2012/732975] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/09/2012] [Indexed: 01/19/2023] Open
Abstract
The hypothalamus is an essential relay in the neural circuitry underlying energy metabolism that needs to continually adapt to changes in the energetic environment. The neuroendocrine control of food intake and energy expenditure is associated with, and likely dependent upon, hypothalamic plasticity. Severe disturbances in energy metabolism, such as those that occur in obesity, are therefore likely to be associated with disruption of hypothalamic transcriptomic plasticity. In this paper, we investigated the effects of two well-characterized antiaging interventions, caloric restriction and voluntary wheel running, in two distinct physiological paradigms, that is, diabetic (db/db) and nondiabetic wild-type (C57/Bl/6) animals to investigate the contextual sensitivity of hypothalamic transcriptomic responses. We found that, both quantitatively and qualitatively, caloric restriction and physical exercise were associated with distinct transcriptional signatures that differed significantly between diabetic and non-diabetic mice. This suggests that challenges to metabolic homeostasis regulate distinct hypothalamic gene sets in diabetic and non-diabetic animals. A greater understanding of how genetic background contributes to hypothalamic response mechanisms could pave the way for the development of more nuanced therapeutics for the treatment of metabolic disorders that occur in diverse physiological backgrounds.
Collapse
Affiliation(s)
- Alexis M. Stranahan
- Physiology Department, Georgia Health Sciences University, Augusta, GA 30912, USA
- *Alexis M. Stranahan:
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Wayne Chadwick
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Sung-Soo Park
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Liyun Wang
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - William H. WoodIII
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| |
Collapse
|
31
|
Abstract
Microglial cells are the resident macrophages in the central nervous system. These cells of mesodermal/mesenchymal origin migrate into all regions of the central nervous system, disseminate through the brain parenchyma, and acquire a specific ramified morphological phenotype termed "resting microglia." Recent studies indicate that even in the normal brain, microglia have highly motile processes by which they scan their territorial domains. By a large number of signaling pathways they can communicate with macroglial cells and neurons and with cells of the immune system. Likewise, microglial cells express receptors classically described for brain-specific communication such as neurotransmitter receptors and those first discovered as immune cell-specific such as for cytokines. Microglial cells are considered the most susceptible sensors of brain pathology. Upon any detection of signs for brain lesions or nervous system dysfunction, microglial cells undergo a complex, multistage activation process that converts them into the "activated microglial cell." This cell form has the capacity to release a large number of substances that can act detrimental or beneficial for the surrounding cells. Activated microglial cells can migrate to the site of injury, proliferate, and phagocytose cells and cellular compartments.
Collapse
|
32
|
Seki T, Fukamizu A, Kiso Y, Mukai H. Mitocryptide-2, a neutrophil-activating cryptide, is a specific endogenous agonist for formyl-peptide receptor-like 1. Biochem Biophys Res Commun 2011; 404:482-7. [DOI: 10.1016/j.bbrc.2010.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/02/2010] [Indexed: 10/18/2022]
|
33
|
Cattaneo F, Guerra G, Ammendola R. Expression and signaling of formyl-peptide receptors in the brain. Neurochem Res 2010; 35:2018-26. [PMID: 21042851 DOI: 10.1007/s11064-010-0301-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2010] [Indexed: 01/05/2023]
Abstract
The human formyl-peptide receptor (FPR) and its variants FPRL1 and FPRL2 belong to the G-protein coupled seven transmembrane receptor (GPCR) family sensitive to pertussis toxin. FPR and FPRL1 were first detected in phagocytic leukocytes, and FPRL2 was found in monocytes and in dendritic cells. The three receptors were subsequently identified in other cell types or tissues, including neuronal cells and brain, where FPR and FPRL1 play a key role in angiogenesis, cell proliferation, protection against and cell death, as well as in neuroendocrine functions. Binding of different agonists to FPRs triggers several signaling pathways, activates NFkB and STAT3 transcriptional factors and induces the accumulation of the CDK inhibitors p21(waf1/cip1), p16(INK4) and p27(kip1). Signaling molecules, such as ERKs, JNK, PKC, p38MAPK, PLC and PLD are involved in these intracellular cascades. In this article we briefly review FPRs expression and signaling in neuronal cells.
Collapse
Affiliation(s)
- Fabio Cattaneo
- Dipartimento di Biochimica e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S Pansini 5, 80131 Naples, Italy
| | | | | |
Collapse
|
34
|
Lee HY, Kim SD, Shim JW, Kim HJ, Kwon JY, Kim JM, Baek SH, Park JS, Bae YS. Activation of human monocytes by a formyl peptide receptor 2-derived pepducin. FEBS Lett 2010; 584:4102-8. [DOI: 10.1016/j.febslet.2010.08.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 08/25/2010] [Accepted: 08/25/2010] [Indexed: 12/22/2022]
|
35
|
Cilibrizzi A, Quinn MT, Kirpotina LN, Schepetkin IA, Holderness J, Ye RD, Rabiet MJ, Biancalani C, Cesari N, Graziano A, Vergelli C, Pieretti S, Dal Piaz V, Giovannoni MP. 6-methyl-2,4-disubstituted pyridazin-3(2H)-ones: a novel class of small-molecule agonists for formyl peptide receptors. J Med Chem 2010; 52:5044-57. [PMID: 19639995 DOI: 10.1021/jm900592h] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Following a ligand-based drug design approach, a potent mixed formyl peptide receptor 1 (FPR1) and formyl peptide receptor-like 1 (FPRL1) agonist (14a) and a potent and specific FPRL1 agonist (14x) were identified. These compounds belong to a large series of pyridazin-3(2H)-one derivatives substituted with a methyl group at position 6 and a methoxy benzyl at position 4. At position 2, an acetamide side chain is essential for activity. Likewise, the presence of lipophilic and/or electronegative substituents in the position para to the aryl group at the end of the chain plays a critical role for activity. Affinity for FPR1 receptors was evaluated by measuring intracellular calcium flux in HL-60 cells transfected with FPR1, FPRL1, and FPRL2. Agonists were able to activate intracellular calcium mobilization and chemotaxis in human neutrophils. The most potent chemotactic agent (EC(50) = 0.6 microM) was the mixed FPR/FPRL1 agonist 14h.
Collapse
Affiliation(s)
- Agostino Cilibrizzi
- Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Firenze, Via Ugo Schiff 6, Sesto Fiorentino 50019 Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Are formyl peptide receptors novel targets for therapeutic intervention in ischaemia-reperfusion injury? Trends Pharmacol Sci 2010; 31:266-76. [PMID: 20483490 PMCID: PMC7112865 DOI: 10.1016/j.tips.2010.04.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 04/06/2010] [Accepted: 04/06/2010] [Indexed: 01/13/2023]
Abstract
Ischaemia–reperfusion (I/R) injury is a common feature of several diseases associated with high morbidity and mortality, such as stroke and myocardial infarction. The damaged tissue displays cardinal signs of inflammation and microvascular injury that, unless resolved, lead to long-term tissue damage with associated dysfunction. Current therapies are limited and are often associated with many side effects. Increasing evidence suggests that members of the formyl peptide receptor (FPR) family, in particular human FPR2/ALX, might have an important role in the pathophysiology of I/R injury. It was recently demonstrated that several peptides and non-peptidyl small-molecule compounds have anti-inflammatory and pro-resolving properties via their action on members of the FPR family. Here I review this evidence and suggest that FPR ligands, particularly in the brain, could be novel and exciting anti-inflammatory therapeutics for the treatment of a variety of clinical conditions, including stroke.
Collapse
|
37
|
Bestebroer J, de Haas CJ, van Strijp JA. How microorganisms avoid phagocyte attraction. FEMS Microbiol Rev 2010; 34:395-414. [DOI: 10.1111/j.1574-6976.2009.00202.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
38
|
Ye N, Wang MW, Qin J, Lin B. Microfluidic devices for characterizing the agonist of formyl peptide receptor in RBL-FPR cells. Biomed Microdevices 2010; 12:513-21. [DOI: 10.1007/s10544-010-9408-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
39
|
Karlsson J, Bylund J, Movitz C, Björkman L, Forsman H, Dahlgren C. A methodological approach to studies of desensitization of the formyl peptide receptor: Role of the read out system, reactive oxygen species and the specific agonist used to trigger neutrophils. J Immunol Methods 2010; 352:45-53. [DOI: 10.1016/j.jim.2009.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 10/22/2009] [Accepted: 10/26/2009] [Indexed: 01/22/2023]
|
40
|
Prat C, Haas PJ, Bestebroer J, de Haas CJC, van Strijp JAG, van Kessel KPM. A homolog of formyl peptide receptor-like 1 (FPRL1) inhibitor from Staphylococcus aureus (FPRL1 inhibitory protein) that inhibits FPRL1 and FPR. THE JOURNAL OF IMMUNOLOGY 2009; 183:6569-78. [PMID: 19846866 DOI: 10.4049/jimmunol.0801523] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The members of the formyl peptide receptor (FPR) family are involved in the sensing of chemoattractant substances, including bacteria-derived N-formylated peptides and host-derived peptides and proteins. We have recently described two chemoattractant receptor inhibitors from Staphylococcus aureus. Chemotaxis inhibitory protein of S. aureus (CHIPS) blocks the formyl peptide receptor (FPR) and the receptor for complement C5a (C5aR), while FPR-like 1 (FPRL1) inhibitory protein (FLIPr) blocks the FPRL1. Here, we describe another staphylococcal chemoattractant-inhibiting protein with 73% overall homology to FLIPr and identical first 25 aa, which we termed FLIPr-like. This protein inhibits neutrophil calcium mobilization and chemotaxis induced by the FPRL1-ligand MMK-1 and FPR-ligand fMLP. While its FPRL1-inhibitory activity lies in the comparable nanomolar range of FLIPr, its antagonism of the FPR is approximately 100-fold more potent than that of FLIPr and comparable to that of CHIPS. The second N-terminal phenylalanine was required for its inhibition of the FPR, but it was dispensable for the FPRL1. Furthermore, the deletion of the first seven amino acids reduced its antagonism of the FPRL1, and the exchange of the first six amino acids with that of CHIPS-conferred receptor specificity. Finally, studies with cells transfected with several chemoattractant receptors confirmed that FLIPr-like specifically binds to the FPR and FPRL1. In conclusion, the newly described excreted protein from S. aureus, FLIPr-like, is a potent inhibitor of the FPR- and FPRL1-mediated neutrophil responses and may be used to selectively modulate these chemoattractant receptors.
Collapse
Affiliation(s)
- Cristina Prat
- Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | | | |
Collapse
|
41
|
Wilms H, Rosenstiel P, Romero-Ramos M, Arlt A, Schäfer H, Seegert D, Kahle P, Odoy S, Claasen J, Holzknecht C, Brandenburg L, Deuschl G, Schreiber S, Kirik D, Lucius R. Suppression of Map Kinases Inhibits Microglial Activation and Attenuates Neuronal Cell Death Induced by α-Synuclein Protofibrils. Int J Immunopathol Pharmacol 2009; 22:897-909. [DOI: 10.1177/039463200902200405] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
α-Synuclein (α-Syn) accounts, as a major component of Lewy bodies (LB), for the filamentous deposits in many cases of neurodegenerative diseases. Yet, little is known about the molecular mechanisms of neuronal loss in these diseases. The correlation between α-Syn oligomerization/aggregation and pathologies raises the key question of which molecular form of α-Syn (i.e. monomeric α-Syn, protofibrils or mature fibrils) represents the damage-inducing culprit in the scenario of synucleinopathies. We show that human α-Syn protofibrils (PFs) are potent activators of parallel proinflammatory signalling pathways (p38 and ERK1/2 MAP kinases and NF-κB) in microglial cells in vitro. Furthermore, stereotactic injection of α-Syn PFs into the substantia nigra of adult rats leads to a profound activation of microglia and adjacent neuronal cell loss, which can be attenuated by the MAP kinase inhibitor semapimod. We propose that the neurodegenerative process of α-synucleinopathies involves microglial activation through α-Syn released or extruded from cells with pathogenic α-Syn metabolism. Compounds that inhibit the MAPK/NF-κB pathways might be a promising pharmacological strategy for the treatment of the inflammatory component of synucleinopathies including PD.
Collapse
Affiliation(s)
| | - P. Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Sweden
| | | | - A. Arlt
- Department of General Internal Medicine, University Hospital Schleswig-Holstein, Campus Kiel
| | - H. Schäfer
- Department of General Internal Medicine, University Hospital Schleswig-Holstein, Campus Kiel
| | | | - P.J. Kahle
- Adolf Butenandt Institute, LMU München
- present address: Laboratory of Functional Neurogenetics, University Clinics Tübingen
| | - S. Odoy
- Adolf Butenandt Institute, LMU München
| | - J.H. Claasen
- Institute of Anatomy, Christian-Albrechts-University Kiel, Campus Kiel, Germany
| | - C. Holzknecht
- Institute of Anatomy, Christian-Albrechts-University Kiel, Campus Kiel, Germany
| | - L.O. Brandenburg
- Institute of Anatomy, Christian-Albrechts-University Kiel, Campus Kiel, Germany
| | - G. Deuschl
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | | | - D. Kirik
- Wallenberg Neuroscience Center, Lund University, Sweden
| | - R. Lucius
- Institute of Anatomy, Christian-Albrechts-University Kiel, Campus Kiel, Germany
| |
Collapse
|
42
|
Abstract
The resolution of inflammation is an active process controlled by endogenous mediators with selective actions on neutrophils and monocytes. The initial phase of the acute inflammatory response is characterized by the production of pro-inflammatory mediators followed by a second phase in which lipid mediators with pro-resolution activities may be generated. The identification of these mediators has provided evidence for the dynamic regulation of the resolution of inflammation. Among these endogenous local mediators of resolution, lipoxins (LXs), lipid mediators typically formed during cell-cell interaction, were the first to be recognized. More recently, families of endogenous chemical mediators, termed resolvins and protectins, were discovered. LXs and aspirin-triggered LXs are considered to act as 'braking signals' in inflammation, limiting the trafficking of leukocytes to the inflammatory site. LXs are actively involved in the resolution of inflammation stimulating non-phlogistic phagocytosis of apoptotic cells by macrophages. Furthermore, LXs have emerged as potential anti-fibrotic mediators that may influence pro-fibrotic cytokines and matrix-associated gene expression in response to growth factors. Here, we provide a review and an update of the biosynthesis, metabolism and bioactions of LXs and LX analogues, and the recent studies on their therapeutic potential as promoters of resolution and fibro-suppressants.
Collapse
Affiliation(s)
- Paola Maderna
- UCD Diabetes Research Centre, UCD Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
43
|
Sun J. Pathogenic Bacterial Proteins and their Anti-Inflammatory Effects in the Eukaryotic Host. Antiinflamm Antiallergy Agents Med Chem 2009; 8:214-227. [PMID: 20090866 DOI: 10.2174/187152309789151986] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bacteria use multiple strategies to bypass the inflammatory responses in order to survive in the host cells. In this review, we discuss the mechanism of the bacerial proteins in inhibiting inflammation. We highlight the anti-inflammatory roles of the type three secretion proteins including Salmonella AvrA, Enteropathogenic Escherichia coli Cif, and Yersinia YopJ, Staphylococcus aureus extracellular adherence protein, and Chlamydia proteins. We also discuss the research progress on the structures of these anti-inflammatory bacterial proteins. The current therapeutic methods for diseases, such as inflammatory bowel diseases, sclerosis, lack influence on the course of chronic inflammation and infection. Therefore, based on the molecular mechanism of the anti-inflammatory bacterial proteins and their 3-Dimension structure, we can design new peptides or non-peptidic molecules that serve as anti-inflammatory drugs without the possible side effect of promoting bacterial infection.
Collapse
Affiliation(s)
- Jun Sun
- Department of Medicine, Gastroenterology & Hepatology Division and Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Ave., Rochester, New York 14642, USA
| |
Collapse
|
44
|
de Paulis A, Prevete N, Rossi FW, Rivellese F, Salerno F, Delfino G, Liccardo B, Avilla E, Montuori N, Mascolo M, Staibano S, Melillo RM, D'Argenio G, Ricci V, Romano M, Marone G. Helicobacter pylori Hp(2-20) promotes migration and proliferation of gastric epithelial cells by interacting with formyl peptide receptors in vitro and accelerates gastric mucosal healing in vivo. THE JOURNAL OF IMMUNOLOGY 2009; 183:3761-9. [PMID: 19692643 DOI: 10.4049/jimmunol.0900863] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori-derived peptide RpL1 aa 2-20 (Hp(2-20)) in addition to its antimicrobial action exerts several immunomodulatory effects in eukaryotic cells by interacting with formyl peptide receptors (FPRs). It has recently been shown that activation of FPRs facilitates intestinal epithelial cell restitution. We investigated whether Hp(2-20) induces healing of injured gastric mucosa and assessed the mechanisms underlying any such effect. We investigated the expression of FPRs in two gastric epithelial cell lines (MKN-28 and AGS) at mRNA and protein level. To determine whether FPRs were functional we performed chemotaxis experiments and proliferation assays and studied the Hp(2-20)-activated downstream signaling pathway. The effect of Hp(2-20) on mucosal healing was evaluated in rats after indomethacin-induced injury. Here we show that: (1) FPRs were expressed in both cell lines; (2) Hp(2-20) stimulated migration and proliferation of gastric epithelial cells; (3) this effect was specifically mediated by formyl peptide receptor-like 1 (FPRL1) and FPRL2 and was associated with activation of FPR-related downstream signaling pathways; (4) Hp(2-20) up-regulated the expression and secretion of vascular endothelial growth factor; and (5) Hp(2-20) accelerated healing of rat gastric mucosa after injury brought about by indomethacin at both the macroscopic and microscopic levels. In conclusion, by interacting with FRPL1 and FPRL2, H. pylori-derived Hp(2-20) induces cell migration and proliferation, as well as the expression of vascular endothelial growth factor, thereby promoting gastric mucosal healing. This study provides further evidence of the complexity of the relationship between H. pylori and human gastric mucosa, and it suggests that a bacterial product may be used to heal gastric mucosal injury.
Collapse
Affiliation(s)
- Amato de Paulis
- Divisione di Immunologia Clinica ed Allergologia e Centro Interdipartimentale di Ricerca di Scienze Immunologiche di Base e Cliniche, Università di Napoli Federico II, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gurevich M, Tuller T, Rubinstein U, Or-Bach R, Achiron A. Prediction of acute multiple sclerosis relapses by transcription levels of peripheral blood cells. BMC Med Genomics 2009; 2:46. [PMID: 19624813 PMCID: PMC2725113 DOI: 10.1186/1755-8794-2-46] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 07/22/2009] [Indexed: 11/10/2022] Open
Abstract
Background The ability to predict the spatial frequency of relapses in multiple sclerosis (MS) would enable physicians to decide when to intervene more aggressively and to plan clinical trials more accurately. Methods In the current study our objective was to determine if subsets of genes can predict the time to the next acute relapse in patients with MS. Data-mining and predictive modeling tools were utilized to analyze a gene-expression dataset of 94 non-treated patients; 62 patients with definite MS and 32 patients with clinically isolated syndrome (CIS). The dataset included the expression levels of 10,594 genes and annotated sequences corresponding to 22,215 gene-transcripts that appear in the microarray. Results We designed a two stage predictor. The first stage predictor was based on the expression level of 10 genes, and predicted the time to next relapse with a resolution of 500 days (error rate 0.079, p < 0.001). If the predicted relapse was to occur in less than 500 days, a second stage predictor based on an additional different set of 9 genes was used to give a more accurate estimation of the time till the next relapse (in resolution of 50 days). The error rate of the second stage predictor was 2.3 fold lower than the error rate of random predictions (error rate = 0.35, p < 0.001). The predictors were further evaluated and found effective both for untreated MS patients and for MS patients that subsequently received immunomodulatory treatments after the initial testing (the error rate of the first level predictor was < 0.18 with p < 0.001 for all the patient groups). Conclusion We conclude that gene expression analysis is a valuable tool that can be used in clinical practice to predict future MS disease activity. Similar approach can be also useful for dealing with other autoimmune diseases that characterized by relapsing-remitting nature.
Collapse
Affiliation(s)
- Michael Gurevich
- Multiple Sclerosis Center, Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | | | | | | | | |
Collapse
|
46
|
Strouse JJ, Young SM, Mitchell HD, Ye RD, Prossnitz ER, Sklar LA, Edwards BS. A novel fluorescent cross-reactive formylpeptide receptor/formylpeptide receptor-like 1 hexapeptide ligand. Cytometry A 2009; 75:264-70. [PMID: 19006074 DOI: 10.1002/cyto.a.20670] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Formylpeptide receptors (FPRs) are implicated in a variety of immunological and inflammatory response cascades. Further understanding of FPR-family ligand interactions could play an integral role in biological and therapeutic discovery. Fluorescent reporter ligands for the family are desirable experimental tools for increased understanding of ligand/receptor interactions. The ligand binding affinity and fluorescent reporting activity of the peptide WK(FL)YMVm was explored though use of the high throughput HyperCyt flow cytometric platform. Relative binding affinities of several known FPR and FPRL1 peptide ligands were compared in a duplex assay format. The fluorescent W-peptide ligand, WK(FL)YMVm, proved to be a high-affinity, cross-reactive reporter ligand for the FPR/FPRL1 duplex assay. Ligand specificity was demonstrated for each receptor, with known, selective peptide ligands. The binding site specificity of the reporter ligand was further verified by a fluorescent confocal microscopy internalization experiment. The fluorescent peptide ligand WK(FL)YMVm binds with high affinity to both FPR and FPRL1. The differential affinities of known peptide ligands were observed with the use of this fluorescent probe in high throughput screening flow cytometry.
Collapse
Affiliation(s)
- J Jacob Strouse
- Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, 87131, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Young SM, Bologa CM, Fara D, Bryant BK, Strouse JJ, Arterburn JB, Ye RD, Oprea TI, Prossnitz ER, Sklar LA, Edwards BS. Duplex high-throughput flow cytometry screen identifies two novel formylpeptide receptor family probes. Cytometry A 2009; 75:253-63. [PMID: 18785269 DOI: 10.1002/cyto.a.20645] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Of recent, clinical interest have been two related human G-protein coupled receptors: formylpeptide receptor (FPR), linked to antibacterial inflammation and malignant glioma cell metastasis; and FPR like-1 (FPRL1), linked to chronic inflammation in systemic amyloidosis, Alzheimer's disease, and prion diseases. In association with the National Institutes of Health (NIH) Molecular Library Screening Network, we implemented a flow-cytometry-based high-throughput screening (HTS) approach for identifying selective small molecule FPR and FPRL1 ligands. The screening assay measured the ability of test compounds to competitively displace a high-affinity, fluorescein- labeled peptide ligand from FPR, FPRL1, or both. U937 cells expressing FPR and rat basophil leukemia (RBL) cells expressing FPRL1 were tested together in a "duplex" format. The U937 cells were color coded with red-fluorescent dye allowing their distinction during analysis. Compounds, cells, and fluorescent ligand were sequentially combined (no wash) in 15 microl assay volumes in 384-well plates. Throughput averaged approximately 11 min per plate to analyze approximately 4,000 cells ( approximately 2,000/receptor) in a 2 microl aspirate from each well. In primary single concentration HTS of 24,304 NIH Small Molecule Repository compounds, 253 resulted in inhibition >30% (181 for FPR, 72 for FPRL1) of which 40 had selective binding inhibition constants (K(i)) < or = 4 microM (34 for FPR and 6 for FPRL1). An additional 1,446 candidate compounds were selected by structure-activity-relationship analysis of the hits and screened to identify novel ligands for FPR (3570-0208, K(i) = 95 +/- 10 nM) and FPRL1 (BB-V-115, K(i) = 270 +/- 51 nM). Each was a selective antagonist in calcium response assays and the most potent small molecule antagonist reported for its respective receptor to date. The duplex assay format reduced assay time, minimized reagent requirements, and provided selectivity information at every screening stage, thus proving to be an efficient means to screen for selective receptor ligand probes.
Collapse
|
48
|
Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev 2009; 61:119-61. [PMID: 19498085 DOI: 10.1124/pr.109.001578] [Citation(s) in RCA: 600] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a small group of seven-transmembrane domain, G protein-coupled receptors that are expressed mainly by mammalian phagocytic leukocytes and are known to be important in host defense and inflammation. The three human FPRs (FPR1, FPR2/ALX, and FPR3) share significant sequence homology and are encoded by clustered genes. Collectively, these receptors bind an extraordinarily numerous and structurally diverse group of agonistic ligands, including N-formyl and nonformyl peptides of different composition, that chemoattract and activate phagocytes. N-formyl peptides, which are encoded in nature only by bacterial and mitochondrial genes and result from obligatory initiation of bacterial and mitochondrial protein synthesis with N-formylmethionine, is the only ligand class common to all three human receptors. Surprisingly, the endogenous anti-inflammatory peptide annexin 1 and its N-terminal fragments also bind human FPR1 and FPR2/ALX, and the anti-inflammatory eicosanoid lipoxin A4 is an agonist at FPR2/ALX. In comparison, fewer agonists have been identified for FPR3, the third member in this receptor family. Structural and functional studies of the FPRs have produced important information for understanding the general pharmacological principles governing all leukocyte chemoattractant receptors. This article aims to provide an overview of the discovery and pharmacological characterization of FPRs, to introduce an International Union of Basic and Clinical Pharmacology (IUPHAR)-recommended nomenclature, and to discuss unmet challenges, including the mechanisms used by these receptors to bind diverse ligands and mediate different biological functions.
Collapse
Affiliation(s)
- Richard D Ye
- Department of Pharmacology, University of Illinois College of Medicine, 835 South Wolcott Avenue, M/C 868, Chicago, Illinois 60612, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhou H, Zhou X, Kouadir M, Zhang Z, Yin X, Yang L, Zhao D. Induction of macrophage migration by neurotoxic prion protein fragment. J Neurosci Methods 2009; 181:1-5. [PMID: 19447501 DOI: 10.1016/j.jneumeth.2009.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 04/02/2009] [Accepted: 04/03/2009] [Indexed: 01/07/2023]
Abstract
Prion diseases are characterized by accumulation of protease resistant isoforms of prion protein (PrP), and infiltration and activation of mononuclear phagocytes at the brain lesions. Interactions between prion proteins and immune cells during disease progression are still not very well understood. In the present study, multiwell chamber chemotaxis assay was carried out to assess the migratory response of macrophage cell line Ana-1 to a synthetic peptide homologous to residues 106-126 of the human prion protein. Specific protein kinase inhibitors were used to elucidate the signaling events underlying PrP106-126-induced macrophages migration, and a comparison with the signaling pattern of macrophage migration induced by substance P (SP) and N-formyl-methionyl-leucyl-phenylalanine (fMLP), respectively, was carried out. The results showed that PrP106-126 had a potent chemotactic effect on murine macrophage cell line Ana-1; that multiple signaling pathways might be involved in the PrP106-126-induced macrophage migrations; and that PrP106-126-induced chemotactic activity was similar to that induced by SP. These findings provide new insights into the mechanisms underlying the interaction between PrP and macrophages.
Collapse
Affiliation(s)
- Haiyun Zhou
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
50
|
Karlsson J, Stenfeldt AL, Rabiet MJ, Bylund J, Forsman HF, Dahlgren C. The FPR2-specific ligand MMK-1 activates the neutrophil NADPH-oxidase, but triggers no unique pathway for opening of plasma membrane calcium channels. Cell Calcium 2009; 45:431-8. [PMID: 19282028 DOI: 10.1016/j.ceca.2009.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 01/29/2009] [Accepted: 02/09/2009] [Indexed: 01/12/2023]
Abstract
Human neutrophils express formyl peptide receptor 1 and 2 (FPR1 and FPR2), two highly homologous G-protein-coupled cell surface receptors important for the cellular recognition of chemotactic peptides. They share many functional as well as signal transduction features, but some fundamental differences have been described. One such difference was recently presented when the FPR2-specific ligand MMK-1 was shown to trigger a unique signal in neutrophils [S. Partida-Sanchez, P. Iribarren, M.E. Moreno-Garcia, et al., Chemotaxis and calcium responses of phagocytes to formyl peptide receptor ligands is differentially regulated by cyclic ADP ribose, J. Immunol. 172 (2004) 1896-1906]. This signal bypassed the emptying of the intracellular calcium stores, a route normally used to open the store-operated calcium channels present in the plasma membrane of neutrophils. Instead, the binding of MMK-1 to FPR2 was shown to trigger a direct opening of the plasma membrane channels. In this report, we add MMK-1 to a large number of FPR2 ligands that activate the neutrophil superoxide-generating NADPH-oxidase. In contrast to earlier findings we show that the transient rise in intracellular free calcium induced by MMK-1 involves both a release of calcium from intracellular stores and an opening of channels in the plasma membrane. The same pattern was obtained with another characterized FPR2 ligand, WKYMVM, and it is also obvious that the two formyl peptide receptor family members trigger the same type of calcium response in human neutrophils.
Collapse
Affiliation(s)
- Jennie Karlsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Sweden.
| | | | | | | | | | | |
Collapse
|