Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 584
Filtrar
1.
Pharmacol Res ; 202: 107125, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38438091

RESUMEN

G protein-coupled receptors (GPCRs) are currently the most widely focused drug targets in the clinic, exerting their biological functions by binding to chemicals and activating a series of intracellular signaling pathways. Formyl-peptide receptor 1 (FPR1) has a typical seven-transmembrane structure of GPCRs and can be stimulated by a large number of endogenous or exogenous ligands with different chemical properties, the first of which was identified as formyl-methionine-leucyl-phenylalanine (fMLF). Through receptor-ligand interactions, FPR1 is involved in inflammatory response, immune cell recruitment, and cellular signaling regulation in key cell types, including neutrophils, neural stem cells (NSCs), and microglia. This review outlines the critical roles of FPR1 in a variety of heart and brain diseases, including myocardial infarction (MI), ischemia/reperfusion (I/R) injury, neurodegenerative diseases, and neurological tumors, with particular emphasis on the milestones of FPR1 agonists and antagonists. Therefore, an in-depth study of FPR1 contributes to the research of innovative biomarkers, therapeutic targets for heart and brain diseases, and clinical applications.


Asunto(s)
Encefalopatías , Receptores de Formil Péptido , Humanos , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Receptores de Formil Péptido/metabolismo , Encéfalo/metabolismo
2.
Front Immunol ; 14: 1112196, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36891309

RESUMEN

Sepsis is defined as a life-threatening dysfunction due to a dysregulated host response to infection. It is a common and complex syndrome and is the leading cause of death in intensive care units. The lungs are most vulnerable to the challenge of sepsis, and the incidence of respiratory dysfunction has been reported to be up to 70%, in which neutrophils play a major role. Neutrophils are the first line of defense against infection, and they are regarded as the most responsive cells in sepsis. Normally, neutrophils recognize chemokines including the bacterial product N-formyl-methionyl-leucyl-phenylalanine (fMLP), complement 5a (C5a), and lipid molecules Leukotriene B4 (LTB4) and C-X-C motif chemokine ligand 8 (CXCL8), and enter the site of infection through mobilization, rolling, adhesion, migration, and chemotaxis. However, numerous studies have confirmed that despite the high levels of chemokines in septic patients and mice at the site of infection, the neutrophils cannot migrate to the proper target location, but instead they accumulate in the lungs, releasing histones, DNA, and proteases that mediate tissue damage and induce acute respiratory distress syndrome (ARDS). This is closely related to impaired neutrophil migration in sepsis, but the mechanism involved is still unclear. Many studies have shown that chemokine receptor dysregulation is an important cause of impaired neutrophil migration, and the vast majority of these chemokine receptors belong to the G protein-coupled receptors (GPCRs). In this review, we summarize the signaling pathways by which neutrophil GPCR regulates chemotaxis and the mechanisms by which abnormal GPCR function in sepsis leads to impaired neutrophil chemotaxis, which can further cause ARDS. Several potential targets for intervention are proposed to improve neutrophil chemotaxis, and we hope that this review may provide insights for clinical practitioners.


Asunto(s)
Síndrome de Dificultad Respiratoria , Sepsis , Animales , Ratones , Neutrófilos/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo
3.
Biochem Pharmacol ; 201: 115091, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35569521

RESUMEN

The over-activated neutrophils through G-protein-coupled receptors (GPCRs) caused inflammation or tissue damage. Therefore, GPCRs or their downstream molecules are major targets for inhibiting uncontrolled neutrophil activation. Our studies investigate the action and underlying mechanism of larixol, a diterpene extract from the root of euphorbia formosana, on fMLP-induced neutrophil respiratory burst, chemotaxis, and granular release. The immunoprecipitation assay was performed to investigate whether larixol inhibits fMLP-induced respiratory burst by interfering with the interaction of fMLP receptor Gi-protein ßγ subunits with its downstream molecules. Briefly, larixol inhibited fMLP (0.1 µM)-induced superoxide anion production (IC50:1.98 ± 0.14 µM), the release of cathepsin G (IC50:2.76 ± 0.15 µM) and chemotaxis in a concentration-dependent manner; however, larixol did not inhibit these functions induced by PMA (100 nM). Larixol inhibited fMLP-induced Src kinase phosphorylation. Therefore, larixol attenuated the downstream signaling of Src kinases, ERK1/2, p38, and AKT phosphorylation. Moreover, larixol inhibited fMLP-induced intracellular calcium mobilization, PKC phosphorylation, and p47phox translocation from the cytosol to the plasma membrane. Larixol inhibited the interaction of the ßγ subunits of Gi-protein of fMLP receptor with Src kinase or with PLCß by the immunoprecipitation and duolink assay. Furthermore, larixol did not antagonize the formyl peptide receptors. Larixol did not increase cyclic nucleotide levels in neutrophils. These results suggest that larixol modulated fMLP-induced neutrophils superoxide anion production, chemotaxis, and granular releases by interrupting the interaction of the ßγ subunits of Gi-protein with downstream signaling of the fMLP receptor.


Asunto(s)
Diterpenos , Receptores de Formil Péptido , Quimiotaxis , Humanos , N-Formilmetionina Leucil-Fenilalanina/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacología , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Fosforilación , Receptores de Formil Péptido/metabolismo , Superóxidos/metabolismo , Familia-src Quinasas/metabolismo
4.
Front Immunol ; 12: 670763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177910

RESUMEN

We collected peripheral blood from thirty-nine elite male endurance runners at rest (24 hours after the last exercise session) and used the Allergy Questionnaire for Athletes score and plasma specific IgE level to separate them into atopic and non-atopic athletes. Neutrophils obtained from atopic and non-atopic athletes were subsequently stimulated in vitro with fMLP (N-formyl-methionyl-leucyl-phenylalanine), LPS (lipopolysaccharide), or PMA (phorbol 12-myristate 13-acetate). Neutrophils from non-atopic runners responded appropriately to LPS, as evidenced by the production of pro (IL-8, TNF-α, and IL-6) and anti-inflammatory (IL-10) cytokines. Neutrophils from atopic elite runners exhibited lower responses to LPS stimulus as indicated by no increase in IL-1ß, TNF-α, and IL-6 production. Neutrophils from non-atopic and atopic runners responded similarly to fMLP stimulation, indicating that migration function remained unaltered. Both groups were unresponsive to PMA induced reactive oxygen species (ROS) production. Training hours and training volume were not associated with neutrophil IgE receptor gene expression or any evaluated neutrophil function. Since non-atopic runners normally responded to LPS stimulation, the reduced neutrophil response to the stimuli was most likely due to the atopic state and not exercise training. The findings reported are of clinical relevance because atopic runners exhibit a constant decline in competition performance and are more susceptible to invading microorganisms.


Asunto(s)
Hipersensibilidad Inmediata/inmunología , Neutrófilos/inmunología , Adulto , Células Cultivadas , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/genética , Infecciones , Lipopolisacáridos/metabolismo , Masculino , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Activación Neutrófila , Resistencia Física , Carrera , Encuestas y Cuestionarios
5.
Cells ; 10(5)2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-34062964

RESUMEN

A malfunction of the innate immune response in COVID-19 is associated with eosinopenia, particularly in more severe cases. This study tested the hypothesis that this eosinopenia is COVID-19 specific and is associated with systemic activation of eosinophils. Blood of 15 healthy controls and 75 adult patients with suspected COVID-19 at the ER were included before PCR testing and analyzed by point-of-care automated flow cytometry (CD10, CD11b, CD16, and CD62L) in the absence or presence of a formyl peptide (fNLF). Forty-five SARS-CoV-2 PCR positive patients were grouped based on disease severity. PCR negative patients with proven bacterial (n = 20) or other viral (n = 10) infections were used as disease controls. Eosinophils were identified with the use of the FlowSOM algorithm. Low blood eosinophil numbers (<100 cells/µL; p < 0.005) were found both in patients with COVID-19 and with other infectious diseases, albeit less pronounced. Two discrete eosinophil populations were identified in healthy controls both before and after activation with fNLF based on the expression of CD11b. Before activation, the CD11bbright population consisted of 5.4% (CI95% = 3.8, 13.4) of total eosinophils. After activation, this population of CD11bbright cells comprised nearly half the population (42.21%, CI95% = 35.9, 54.1). Eosinophils in COVID-19 had a similar percentage of CD11bbright cells before activation (7.6%, CI95% = 4.5, 13.6), but were clearly refractory to activation with fNLF as a much lower percentage of cells end up in the CD11bbright fraction after activation (23.7%, CI95% = 18.5, 27.6; p < 0.001). Low eosinophil numbers in COVID-19 are associated with refractoriness in responsiveness to fNLF. This might be caused by migration of fully functional cells to the tissue.


Asunto(s)
COVID-19/inmunología , Eosinófilos/inmunología , Inmunidad Innata , N-Formilmetionina Leucil-Fenilalanina/metabolismo , SARS-CoV-2/inmunología , Adulto , COVID-19/sangre , COVID-19/diagnóstico , COVID-19/virología , Prueba de Ácido Nucleico para COVID-19 , Estudios de Casos y Controles , Separación Celular , Estudios de Cohortes , Eosinófilos/metabolismo , Citometría de Flujo , Voluntarios Sanos , Humanos , Recuento de Leucocitos , ARN Viral/aislamiento & purificación , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad
6.
J Immunol ; 205(9): 2545-2553, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32938725

RESUMEN

Pharmacological activation of integrin CD11b/CD18 (αMß2, Mac-1, and CR3) shows anti-inflammatory benefits in a variety of animal models of human disease, and it is a novel therapeutic strategy. Reasoning that genetic models can provide an orthogonal and direct system for the mechanistic study of CD11b agonism, we present in this study, to our knowledge, a novel knock-in model of constitutive active CD11b in mice. We genetically targeted the Itgam gene (which codes for CD11b) to introduce a point mutation that results in the I332G substitution in the protein. The I332G mutation in CD11b promotes an active, higher-affinity conformation of the ligand-binding I/A-domain (CD11b αA-domain). In vitro, this mutation increased adhesion of knock-in neutrophils to fibrinogen and decreased neutrophil chemotaxis to a formyl-Met-Leu-Phe gradient. In vivo, CD11bI332G animals showed a reduction in recruitment of neutrophils and macrophages in a model of sterile peritonitis. This genetic activation of CD11b also protected against development of atherosclerosis in the setting of hyperlipidemia via reduction of macrophage recruitment into atherosclerotic lesions. Thus, our animal model of constitutive genetic activation of CD11b can be a useful tool for the study of integrin activation and its potential contribution to modulating leukocyte recruitment and alleviating different inflammatory diseases.


Asunto(s)
Antígeno CD11b/genética , Antígenos CD18/genética , Integrinas/genética , Animales , Adhesión Celular/genética , Quimiotaxis de Leucocito/genética , Modelos Animales de Enfermedad , Femenino , Fibrinógeno/genética , Leucocitos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , N-Formilmetionina Leucil-Fenilalanina/análogos & derivados , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/metabolismo
7.
Nat Commun ; 11(1): 1208, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32139677

RESUMEN

The human formyl peptide receptor 2 (FPR2) plays a crucial role in host defense and inflammation, and has been considered as a drug target for chronic inflammatory diseases. A variety of peptides with different structures and origins have been characterized as FPR2 ligands. However, the ligand-binding modes of FPR2 remain elusive, thereby limiting the development of potential drugs. Here we report the crystal structure of FPR2 bound to the potent peptide agonist WKYMVm at 2.8 Å resolution. The structure adopts an active conformation and exhibits a deep ligand-binding pocket. Combined with mutagenesis, ligand binding and signaling studies, key interactions between the agonist and FPR2 that govern ligand recognition and receptor activation are identified. Furthermore, molecular docking and functional assays reveal key factors that may define binding affinity and agonist potency of formyl peptides. These findings deepen our understanding about ligand recognition and selectivity mechanisms of the formyl peptide receptor family.


Asunto(s)
Receptores de Formil Péptido/química , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/química , Receptores de Lipoxina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Mutación/genética , N-Formilmetionina Leucil-Fenilalanina/química , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
8.
Sci Rep ; 9(1): 18540, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31811262

RESUMEN

Eugenol is a polyphenol extracted from Syzygium aromaticum essential oil. It is known to have anti-inflammatory and chemoprotective properties as well as a potent anti-oxidant activity due the presence of its phenolic group. In this study, we examined the effects of eugenol on neutrophil superoxide production, a key process involved in innate immunity and inflammation. Superoxide anion generationin human neutrophils was measured by cytochrome c reduction assay. Western blotting was used to analyze the phosphorylation of, p47phox, MAPKinases (p38 and ERK1/2), MEK1/2 and Raf, key proteins involved in the activation of NADPH oxidase. Pretreatment of neutrophils by increasing concentrations (2.5 µg/mL-20 µg/mL) of eugenol for 30 min, inhibited significantly (p < 0.001) superoxide anion generation induced by the chemotactic peptide formyl-Met-Leu-Phe (fMLF) with an IC50 of 5 µg/mL. Phorbolmyristate acetate (PMA)-stimulated O2- production was affected only at the highest eugenol concentration (20 µg/mL). Results showed that eugenol decreased the phosphorylation of p47phox onSer-345 and Ser-328, the translocation of p47phox to the membranesand the phosphorylation of Raf, MEK1/2 and ERK1/2 proteins. Taken together, our results suggest that eugenol inhibits the generation of superoxide anion by neutrophils via the inhibition of Raf/MEK/ERK1/2/p47phox-phosphorylation pathway.


Asunto(s)
Antioxidantes/farmacología , Eugenol/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , NADPH Oxidasas/metabolismo , Neutrófilos/efectos de los fármacos , Quimiotaxis/inmunología , Voluntarios Sanos , Humanos , Sistema de Señalización de MAP Quinasas/inmunología , N-Formilmetionina Leucil-Fenilalanina/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Superóxidos/metabolismo
9.
Mar Drugs ; 17(8)2019 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-31394844

RESUMEN

Two new capnosane-based diterpenoids, flaccidenol A (1) and 7-epi-pavidolide D (2), two new cembranoids, flaccidodioxide (3) and flaccidodiol (4), and three known compounds 5 to 7 were characterized from the marine soft coral Klyxum flaccidum, collected off the coast of the island of Pratas. The structures of the new compounds were determined by extensive spectroscopic analyses, including 1D and 2D nuclear magnetic resonance (NMR) spectroscopy, and spectroscopic data comparison with related structures. The rare capnosane diterpenoids were isolated herein from the genus Klyxum for the first time. The cytotoxicity of compounds 1 to 7 against the proliferation of a limited panel of cancer cell lines was assayed. The isolated diterpenoids also exhibited anti-inflammatory activity through suppression of superoxide anion generation and elastase release in the N-formyl-methionyl-leucyl-phenylalanine/cytochalasin B (fMLF/CB)-stimulated human neutrophils. Furthermore, 1 and 7 also exhibited cytotoxicity toward the tested cancer cells, and 7 could effectively inhibit elastase release. It is worth noting that the biological activities of 7 are reported for the first time in this paper.


Asunto(s)
Antozoos/química , Factores Biológicos/farmacología , Diterpenos/farmacología , Animales , Antiinflamatorios/farmacología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocalasina B/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Espectroscopía de Resonancia Magnética/métodos , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Elastasa Pancreática/metabolismo , Superóxidos/metabolismo
10.
Nutrients ; 11(6)2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-31212992

RESUMEN

Vitamin C (ascorbate) is important for neutrophil function and immune health. Studies showing improved immune function have primarily used cells from scorbutic animals or from individuals with infectious conditions or immune cell disorders. Few studies have focused on the requirements of neutrophils from healthy adults. Therefore, we have investigated the role of vitamin C, at concentrations equivalent to those obtained in plasma from oral intakes (i.e., 50-200 µmol/L), on key functions of neutrophils isolated from healthy individuals. Cells were either pre-loaded with dehydroascorbic acid, which is rapidly reduced intracellularly to ascorbate, or the cells were activated in the presence of extracellular ascorbate. We measured the effects of enhanced ascorbate uptake on the essential functions of chemotaxis, oxidant production, programmed cell death and neutrophil extracellular trap (NET) formation. We found that neutrophils isolated from healthy individuals already had replete ascorbate status (0.35 nmol/106 cells), therefore they did not uptake additional ascorbate. However, they readily took up dehydroascorbic acid, thus significantly increasing their intracellular ascorbate concentrations, although this was found to have no additional effect on superoxide production or chemotaxis. Interestingly, extracellular ascorbate appeared to enhance directional mobilityin the presence of the chemoattractant formyl-methionyl-leucyl-phenylalanine (fMLP). Stimulation of the cells in the presence of ascorbate significantly increased intracellular ascorbate concentrations and, although this exhibited a non-significant increase in phosphatidylserine exposure, NET formation was significantly attenuated. Our findings demonstrate the ability of neutrophils to regulate their uptake of ascorbate from the plasma of healthy humans to maintain an optimal level within the cell for proper functioning. Higher oral intakes, however, may help reduce tissue damage and inflammatory pathologies associated with NET formation.


Asunto(s)
Ácido Ascórbico/fisiología , Neutrófilos/metabolismo , Transporte Biológico , Quimiotaxis , Ácido Deshidroascórbico/metabolismo , Trampas Extracelulares/metabolismo , Voluntarios Sanos , Humanos , N-Formilmetionina Leucil-Fenilalanina/metabolismo
11.
Front Immunol ; 10: 1270, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244835

RESUMEN

Despite recent advances in our understanding of the mechanisms underlying systemic inflammatory response syndrome (SIRS) and sepsis, the current therapeutic approach to these critically ill patients is centered around supportive care including fluid resuscitation, vasopressors and source control. The incidence of SIRS and sepsis continues to increase in the United States and patients die due to failure to respond to the traditional therapies of nitric oxide blockade, adrenergic agonists, etc. Bacterial and mitochondrial N-formyl peptides (NFPs) act as damage-associated molecular patterns and activate the innate immune system through formyl peptide receptors (FPR) located in immune and non-immune cells, including the vascular endothelium. The resulting inflammatory response manifests as capillary leak, tissue hypoperfusion and vasoplegia, partially due to endothelium barrier breakdown. Potential strategies to prevent this response include decreasing NFP release, breakdown of NFPs, and blocking NFPs from binding FPR. We propose the use of deformylase, the degrading enzyme for NFPs, as potential therapeutic approach to prevent the deleterious effects of NFPs in SIRS and sepsis.


Asunto(s)
Bacteriemia/metabolismo , Bacteriemia/microbiología , Endotelio Vascular/metabolismo , Mitocondrias/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Susceptibilidad a Enfermedades , Desarrollo de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Terapia Molecular Dirigida , Permeabilidad , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/metabolismo , Sepsis/etiología , Sepsis/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo
12.
Mol Pharm ; 16(6): 2636-2647, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31067065

RESUMEN

G protein-coupled receptors (GPCRs) undergo ligand-induced internalization that carries the cognate ligands into intracellular compartments. The present study explores this property for the use of formyl peptide receptor 1 (FPR1), a class A GPCR that binds formylated peptides, as a potential target for drug delivery. A pH-sensitive peptide-drug conjugate consisting of doxorubicin (DOX), N-ε-maleimidocaproic acid hydrazide (EMCH), and the formyl peptide fMet-Leu-Phe-Cys (abbreviated as DEF) was prepared. DEF retained pharmacological activities of formyl peptides in binding to FPR1 and mobilization of Ca2+ from intracellular stores. However, the conjugated DOX was no longer cell membrane-permeable and relied on FPR1 for cellular entry. DOX was released from DEF into acidic compartments labeled with fluorescent trackers for endosomes. Treatment of cells with pharmacological inhibitors that block clathrin- or caveolae-mediated endocytosis did not abrogate FPR1-dependent DEF internalization, nor did inhibition of macropinocytosis and phagocytosis. In contrast, cholesterol depletion abrogated DEF internalization through FPR1, suggesting characteristics of cholesterol-dependent uptake mediated by a cell surface receptor. These results demonstrate the possibility of using FPR1 for targeted drug delivery.


Asunto(s)
Colesterol/metabolismo , Endocitosis/fisiología , Receptores de Formil Péptido/metabolismo , Cromatografía Líquida de Alta Presión , Endocitosis/genética , Células HeLa , Humanos , Espectrometría de Masas , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/metabolismo , Receptores de Formil Péptido/genética
13.
Front Immunol ; 10: 359, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30915068

RESUMEN

Neutrophils are the first responders to infection and play a pivotal role in many inflammatory diseases, including sepsis. Recent studies have shown that lipopolysaccharide (LPS), a classical pattern recognition molecule, dynamically programs innate immune responses. In this study, we show that pre-treatment with super-low levels of LPS [1 ng/mL] significantly dysregulate neutrophil migratory phenotypes, including spontaneous migration and altering neutrophil decision-making. To quantify neutrophil migratory decision-making with single-cell resolution, we developed a novel microfluidic competitive chemotaxis-chip (µC3) that exposes cells in a central channel to competing chemoattractant gradients. In this reductionist approach, we use two chemoattractants: a pro-resolution (N-Formyl-Met-Leu-Phe, fMLP) and pro-inflammatory (Leukotriene B4, LTB4) chemoattractant to model how a neutrophil makes a decision to move toward an end target chemoattractant (e.g., bacterial infection) vs. an intermediary chemoattractant (e.g., inflammatory signal). We demonstrate that naïve neutrophils migrate toward the primary end target signal in higher percentages than toward the secondary intermediary signal. As expected, we found that training with high dose LPS [100 ng/mL] influences a higher percentage of neutrophils to migrate toward the end target signal, while reducing the percentage of neutrophils that migrate toward the intermediary signal. Surprisingly, super-low dose LPS [1 ng/mL] significantly changes the ratios of migrating cells and an increased percentage of cells migrate toward the intermediary signal. Significantly, there was also an increase in the numbers of spontaneously migrating neutrophils after treatment with super-low dose LPS. These results shed light onto the directional migratory decision-making of neutrophils exposed to inflammatory training signals. Understanding these mechanisms may lead to the development of pro-resolution therapies that correct the neutrophil compass and reduce off-target organ damage.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Neutrófilos/inmunología , Supervivencia Celular/efectos de los fármacos , Factores Quimiotácticos/metabolismo , Células HL-60 , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/metabolismo , Dispositivos Laboratorio en un Chip , Leucotrieno B4/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/efectos de los fármacos
14.
JCI Insight ; 3(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30385721

RESUMEN

TGF-ß signals through a receptor complex composed of 2 type I and 2 type II (TGF-ßRII) subunits. We investigated the role of macrophage TGF-ß signaling in fibrosis after AKI in mice with selective monocyte/macrophage TGF-ßRII deletion (macrophage TGF-ßRII-/- mice). Four weeks after injury, renal TGF-ß1 expression and fibrosis were higher in WT mice than macrophage TGF-ßRII-/- mice, which had decreased renal macrophages. The in vitro chemotactic response to f-Met-Leu-Phe was comparable between bone marrow-derived monocytes (BMMs) from WT and macrophage TGF-ßRII-/- mice, but TGF-ßRII-/- BMMs did not respond to TGF-ß. We then implanted Matrigel plugs suffused with either f-Met-Leu-Phe or TGF-ß1 into WT or macrophage TGF-ßRII-/- mice. After 6 days, f-Met-Leu-Phe induced similar macrophage infiltration into the Matrigel plugs of WT and macrophage TGF-ßRII-/- mice, but TGF-ß induced infiltration only in WT mice. We further determined the number of labeled WT or TGF-ßRII-/- BMMs infiltrating into WT kidneys 20 days after ischemic injury. There were more labeled WT BMMs than TGF-ßRII-/- BMMs. Therefore, macrophage TGF-ßRII deletion protects against the development of tubulointerstitial fibrosis following severe ischemic renal injury. Chemoattraction of macrophages to the injured kidney through a TGF-ß/TGF-ßRII axis is a heretofore undescribed mechanism by which TGF-ß can mediate renal fibrosis during progressive renal injury.


Asunto(s)
Lesión Renal Aguda/patología , Fibrosis/metabolismo , Riñón/metabolismo , Macrófagos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Lesión Renal Aguda/complicaciones , Animales , Células de la Médula Ósea/citología , Factores Quimiotácticos/metabolismo , Factores Quimiotácticos/fisiología , Fibrosis/etiología , Riñón/patología , Masculino , Ratones , Ratones Transgénicos/metabolismo , Monocitos/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
15.
J Biol Chem ; 293(33): 12690-12702, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29929985

RESUMEN

Neutrophils are white blood cells that are mobilized to damaged tissues and to sites of pathogen invasion, providing the first line of host defense. Chemokines displayed on the surface of blood vessels promote migration of neutrophils to these sites, and tissue- and pathogen-derived chemoattractant signals, including N-formylmethionylleucylphenylalanine (fMLP), elicit further migration to sites of infection. Although nearly all chemoattractant receptors use heterotrimeric G proteins to transmit signals, many of the mechanisms lying downstream of chemoattractant receptors that either promote or limit neutrophil motility are incompletely defined. Here, we show that regulator of G protein signaling 5 (RGS5), a protein that modulates G protein activity, is expressed in both human and murine neutrophils. We detected significantly more neutrophils in the airways of Rgs5-/- mice than WT counterparts following acute respiratory virus infection and in the peritoneum in response to injection of thioglycollate, a biochemical proinflammatory stimulus. RGS5-deficient neutrophils responded with increased chemotaxis elicited by the chemokines CXC motif chemokine ligand 1 (CXCL1), CXCL2, and CXCL12 but not fMLP. Moreover, adhesion of these cells was increased in the presence of both CXCL2 and fMLP. In summary, our results indicate that RGS5 deficiency increases chemotaxis and adhesion, leading to more efficient neutrophil mobilization to inflamed tissues in mice. These findings suggest that RGS5 expression and activity in neutrophils determine their migrational patterns in the complex microenvironments characteristic of inflamed tissues.


Asunto(s)
Factores Quimiotácticos/metabolismo , Quimiotaxis , Neutrófilos/patología , Proteínas RGS/metabolismo , Proteínas RGS/fisiología , Animales , Adhesión Celular , Movimiento Celular , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/metabolismo , Transducción de Señal
16.
Microbiol Immunol ; 61(12): 523-530, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28976590

RESUMEN

Trehalose 6,6'-dimycolate (TDM), or cord factor, is a crucial stimulus of immune responses during Mycobacterium tuberculosis infection. Although TDM has immuno-stimulatory properties, including adjuvant activity and the ability to induce granuloma formation, the mechanisms underlying these remain unknown. We hypothesized that TDM stimulates transendothelial migration of neutrophils, which are the first immune cells to infiltrate the tissue upon infection. In this study, it was shown that TDM enhances N-formylmethionyl-leucyl-phenylalanine (fMLP)-induced chemotaxis and transendothelial movement by prolonging AKT phosphorylation in human neutrophils. TDM induced expression of macrophage-inducible C-type lectin, a receptor for TDM, and induced secretion of pro-inflammatory cytokines and chemokines in differentiated HL-60 cells. In 2- and 3-D neutrophil migration assays, TDM-stimulated neutrophils showed increased fMLP-induced chemotaxis and transendothelial migration. Interestingly, following fMLP stimulation of TDM-activated neutrophils, AKT, a crucial kinase for neutrophil polarization and chemotaxis, showed prolonged phosphorylation at serine 473. Taken together, these data suggest that TDM modulates transendothelial migration of neutrophils upon mycobacterial infection through prolonged AKT phosphorylation. AKT may therefore be a promising therapeutic target for enhancing immune responses to mycobacterial infection.


Asunto(s)
Movimiento Celular , Factores Cordón/metabolismo , Mycobacterium tuberculosis/metabolismo , Neutrófilos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tuberculosis/enzimología , Secuencias de Aminoácidos , Células HL-60 , Interacciones Huésped-Patógeno , Humanos , Mycobacterium tuberculosis/genética , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/enzimología , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Tuberculosis/genética , Tuberculosis/microbiología , Tuberculosis/fisiopatología
17.
Innate Immun ; 22(8): 635-646, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27655046

RESUMEN

Neutrophil (polymorphonuclear leukocyte) activation with release of granule contents plays an important role in the pathogenesis of acute lung injury, prompting clinical trials of inhibitors of neutrophil elastase. Despite mounting evidence for neutrophil-mediated host tissue damage in a variety of disease processes, mechanisms regulating azurophilic granule exocytosis at the plasma membrane, and thus release of elastase and other proteases, are poorly characterized. We hypothesized that azurophilic granule exocytosis would be enhanced under priming conditions similar to those seen during acute inflammatory events and during chronic inflammatory disease, and selected the cytokine TNF-α to model this in vitro. Neutrophils stimulated with TNF-α alone elicited intracellular reactive oxygen species (ROS) generation and mobilization of secretory vesicles, specific, and gelatinase granules. p38 and ERK1/2 MAPK were involved in these components of priming. TNF-α priming alone did not mobilize azurophilic granules to the cell surface, but did markedly increase elastase release into the extracellular space in response to secondary stimulation with N-formyl-Met-Leu-Phe (fMLF). Priming of fMLF-stimulated elastase release was further augmented in the absence of NADPH oxidase-derived ROS. Our findings provide a mechanism for host tissue damage during neutrophil-mediated inflammation and suggest a novel anti-inflammatory role for the NADPH oxidase.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Elastasa de Leucocito/metabolismo , NADPH Oxidasas/metabolismo , Neutrófilos/inmunología , Vesículas Secretoras/metabolismo , Colorantes Azulados/química , Degranulación de la Célula , Células Cultivadas , Exocitosis , Gelatinasas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Activación Neutrófila , Especies Reactivas de Oxígeno/metabolismo , Vesículas Secretoras/química , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
J Agric Food Chem ; 64(38): 7156-70, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27586050

RESUMEN

Essential oils were obtained by hydrodistillation of the umbels+seeds and stems of Ferula akitschkensis (FAEOu/s and FAEOstm, respectively) and analyzed by gas chromatography and gas chromatography-mass spectrometry. Fifty-two compounds were identified in FAEOu/s; the primary components were sabinene, α-pinene, ß-pinene, terpinen-4-ol, eremophilene, and 2-himachalen-7-ol, whereas the primary components of FAEOstm were myristicin and geranylacetone. FAEOu/s, ß-pinene, sabinene, γ-terpinene, geranylacetone, isobornyl acetate, and (E)-2-nonenal stimulated [Ca(2+)]i mobilization in human neutrophils, with the most potent being geranylacetone (EC50 = 7.6 ± 1.9 µM) and isobornyl acetate 6.4 ± 1.7 (EC50 = 7.6 ± 1.9 µM). In addition, treatment of neutrophils with ß-pinene, sabinene, γ-terpinene, geranylacetone, and isobornyl acetate desensitized the cells to N-formyl-Met-Leu-Phe (fMLF)- and interleukin-8 (IL-8)-induced [Ca(2+)]i flux and inhibited fMLF-induced chemotaxis. The effects of ß-pinene, sabinene, γ-terpinene, geranylacetone, and isobornyl acetate on neutrophil [Ca(2+)]i flux were inhibited by transient receptor potential (TRP) channel blockers. Furthermore, the most potent compound, geranylacetone, activated Ca(2+) influx in TRPV1-transfected HEK293 cells. In contrast, myristicin inhibited neutrophil [Ca(2+)]i flux stimulated by fMLF and IL-8 and inhibited capsaicin-induced Ca(2+) influx in TRPV1-transfected HEK293 cells. These findings, as well as pharmacophore modeling of TRP agonists, suggest that geranylacetone is a TRPV1 agonist, whereas myristicin is a TRPV1 antagonist. Thus, at least part of the medicinal properties of Ferula essential oils may be due to modulatory effects on TRP channels.


Asunto(s)
Ferula/química , Factores Inmunológicos/farmacología , Neutrófilos/efectos de los fármacos , Aceites Volátiles/farmacología , Aceites de Plantas/farmacología , Aldehídos/farmacología , Canfanos/farmacología , Capsaicina/farmacología , Movimiento Celular/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas , Células HEK293 , Células HL-60 , Humanos , Interleucina-8/metabolismo , N-Formilmetionina Leucil-Fenilalanina/análogos & derivados , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/metabolismo , Aceites Volátiles/química , Aceites de Plantas/química , Semillas/química , Canales Catiónicos TRPV/metabolismo , Terpenos/farmacología , Canales de Potencial de Receptor Transitorio/metabolismo
19.
Genes Immun ; 17(6): 321-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27307211

RESUMEN

G Protein Signaling Modulator-3 (GPSM3) is a leukocyte-specific regulator of G protein-coupled receptors (GPCRs), which binds inactivated Gαi·GDP subunits and precludes their reassociation with Gßγ subunits. GPSM3 deficiency protects mice from inflammatory arthritis and, in humans, GPSM3 single-nucleotide polymorphisms (SNPs) are inversely associated with the risk of rheumatoid arthritis development; recently, these polymorphisms were linked to one particular SNP (rs204989) that decreases GPSM3 transcript abundance. However, the precise role of GPSM3 in leukocyte biology is unknown. Here, we show that GPSM3 is induced in the human promyelocytic leukemia NB4 cell line following retinoic acid treatment, which differentiates this cell line into a model of neutrophil physiology (NB4*). Reducing GPSM3 expression in NB4* cells, akin to the effect ascribed to the rs204989 C>T transition, disrupts cellular migration toward leukotriene B4 (LTB4) and (to a lesser extent) interleukin-8 (a.k.a. IL-8 or CXCL8), but not migration toward formylated peptides (fMLP). As the chemoattractants LTB4 and CXCL8 are involved in recruitment of neutrophils to the arthritic joint, our results suggest that the arthritis-protective GPSM3 SNP rs204989 may act to decrease neutrophil chemoattractant responsiveness.


Asunto(s)
Artritis Reumatoide/genética , Quimiotaxis de Leucocito , Inhibidores de Disociación de Guanina Nucleótido/fisiología , Neutrófilos/metabolismo , Artritis Reumatoide/inmunología , Línea Celular Tumoral , Quimiotaxis de Leucocito/genética , Inhibidores de Disociación de Guanina Nucleótido/genética , Humanos , Interleucina-8/metabolismo , Leucopoyesis , Leucotrieno B4/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Polimorfismo de Nucleótido Simple , Tretinoina/metabolismo
20.
Pulm Pharmacol Ther ; 37: 49-56, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26923940

RESUMEN

Respiratory failure is a common characteristic of systemic inflammatory response syndrome (SIRS) and sepsis. Trauma and severe blood loss cause the release of endogenous molecules known as damage-associated molecular patterns (DAMPs). Mitochondrial N-formyl peptides (F-MITs) are DAMPs that share similarities with bacterial N-formylated peptides, and are potent immune system activators. Recently, we observed that hemorrhagic shock-induced increases in plasma levels of F-MITs associated with lung damage, and that antagonism of formyl peptide receptors (FPR) ameliorated hemorrhagic shock-induced lung injury in rats. Corroborating these data, in the present study, it was observed that F-MITs expression is higher in plasma samples from trauma patients with SIRS or sepsis when compared to control trauma group. Therefore, to better understand the role of F-MITs in the regulation of lung and airway function, we studied the hypothesis that F-MITs lead to airway contraction and lung inflammation. We observed that F-MITs induced concentration-dependent contraction in trachea, bronchi and bronchioles. However, pre-treatment with mast cells degranulator or FPR antagonist decreased this response. Finally, intratracheal challenge with F-MITs increased neutrophil elastase expression in lung and inducible nitric oxide synthase and cell division control protein 42 expression in all airway segments. These data suggest that F-MITs could be a putative target to treat respiratory failure in trauma patients.


Asunto(s)
Mitocondrias/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Infiltración Neutrófila/fisiología , Receptores de Formil Péptido/metabolismo , Adolescente , Adulto , Animales , Humanos , Lesión Pulmonar/fisiopatología , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Neutrófilos/metabolismo , Ratas , Ratas Wistar , Insuficiencia Respiratoria/fisiopatología , Sepsis/fisiopatología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...