Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 578, 2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35102146

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is an important health concern worldwide and progresses into nonalcoholic steatohepatitis (NASH). Although prevalence and severity of NAFLD/NASH are higher in men than premenopausal women, it remains unclear how sex affects NAFLD/NASH pathophysiology. Formyl peptide receptor 2 (FPR2) modulates inflammatory responses in several organs; however, its role in the liver is unknown. Here we show that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH. NASH-like liver injury was induced in both sexes during choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) feeding, but compared with females, male mice had more severe hepatic damage. Fpr2 was more highly expressed in hepatocytes and healthy livers from females than males, and FPR2 deletion exacerbated liver damage in CDAHFD-fed female mice. Estradiol induced Fpr2 expression, which protected hepatocytes and the liver from damage. In conclusion, our results demonstrate that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH, suggesting a novel therapeutic target for NAFLD/NASH.


Assuntos
Progressão da Doença , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Receptores de Formil Peptídeo/metabolismo , Caracteres Sexuais , Animais , Biomarcadores/metabolismo , Células Cultivadas , Deficiência de Colina/complicações , Citoproteção/efeitos dos fármacos , Dieta Hiperlipídica , Estradiol/sangue , Estradiol/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Feminino , Deleção de Genes , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Inflamação/patologia , Lipídeos/toxicidade , Lipoproteínas VLDL/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Formil Peptídeo/deficiência , Regulação para Cima/efeitos dos fármacos
2.
Mol Metab ; 31: 138-149, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31918915

RESUMO

OBJECTIVE: Recently, we observed that the specialized proresolving mediator (SPM) entity resolvin D1 activates lipoxin A4/formyl peptide receptor 2 (ALX/FPR2), which facilitates cardiac healing and persistent inflammation is a hallmark of impaired cardiac repair in aging. Splenic leukocyte-directed SPMs are essential for the safe clearance of inflammation and cardiac repair after injury; however, the target of SPMs remains undefined in cardiac healing and repair. METHODS: To define the mechanistic basis of ALX/FPR2 as a resolvin D1 target, ALX/FPR2-null mice were examined extensively. The systolic-diastolic heart function was assessed using echocardiography, leukocytes were phenotyped using flow cytometry, and SPMs were quantitated using mass spectrometry. The presence of cardiorenal syndrome was validated using histology and renal markers. RESULTS: Lack of ALX/FPR2 led to the development of spontaneous obesity and diastolic dysfunction with reduced survival with aging. After cardiac injury, ALX/FPR2-/- mice showed lower expression of lipoxygenases (-5, -12, -15) and a reduction in SPMs in the infarcted left ventricle and spleen, indicating nonresolving inflammation. Reduced SPM levels in the infarcted heart and spleen are suggestive of impaired cross-talk between the injured heart and splenic leukocytes, which are required for the resolution of inflammation. In contrast, cyclooxygenases (-1 and -2) were over amplified in the infarcted heart. Together, these results suggest interorgan signaling in which the spleen acts as both an SPM biosynthesizer and supplier in acute heart failure. ALX/FPR2 dysfunction magnified obesogenic cardiomyopathy and renal inflammation (↑NGAL, ↑TNF-α, ↑CCL2, ↑IL-1ß) with elevated plasma creatinine levels in aging mice. At the cellular level, ALX/FPR2-/- mice showed impairment of macrophage phagocytic function ex-vivo with expansion of neutrophils after myocardial infarction. CONCLUSIONS: Lack of ALX/FPR2 induced obesity, reduced the life span, amplified leukocyte dysfunction, and facilitated profound interorgan nonresolving inflammation. Our study shows the integrative and indispensable role of ALX/FPR2 in lipid metabolism, cardiac inflammation-resolution processes, obesogenic aging, and renal homeostasis.


Assuntos
Insuficiência Cardíaca/metabolismo , Inflamação/metabolismo , Lipoxinas/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Fatores Etários , Animais , Insuficiência Cardíaca/patologia , Humanos , Inflamação/patologia , Lipoxinas/deficiência , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Formil Peptídeo/deficiência , Receptores de Lipoxinas/deficiência
3.
Nature ; 574(7776): 57-62, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31534221

RESUMO

The causative agent of plague, Yersinia pestis, uses a type III secretion system to selectively destroy immune cells in humans, thus enabling Y. pestis to reproduce in the bloodstream and be transmitted to new hosts through fleabites. The host factors that are responsible for the selective destruction of immune cells by plague bacteria are unknown. Here we show that LcrV, the needle cap protein of the Y. pestis type III secretion system, binds to the N-formylpeptide receptor (FPR1) on human immune cells to promote the translocation of bacterial effectors. Plague infection in mice is characterized by high mortality; however, Fpr1-deficient mice have increased survival and antibody responses that are protective against plague. We identified FPR1R190W as a candidate resistance allele in humans that protects neutrophils from destruction by the Y. pestis type III secretion system. Thus, FPR1 is a plague receptor on immune cells in both humans and mice, and its absence or mutation provides protection against Y. pestis. Furthermore, plague selection of FPR1 alleles appears to have shaped human immune responses towards other infectious diseases and malignant neoplasms.


Assuntos
Macrófagos/metabolismo , Neutrófilos/metabolismo , Peste/microbiologia , Receptores de Formil Peptídeo/metabolismo , Yersinia pestis/metabolismo , Alelos , Animais , Antígenos de Bactérias/metabolismo , Aderência Bacteriana , Sistemas CRISPR-Cas , Quimiotaxia/imunologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Peste/imunologia , Peste/prevenção & controle , Polimorfismo de Nucleotídeo Único/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Sistemas de Secreção Tipo III/efeitos dos fármacos , Células U937 , Yersinia pestis/química , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
4.
J Thromb Haemost ; 17(7): 1120-1133, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31033193

RESUMO

Essentials The role of formyl peptide receptor 1 (FPR1) and its ligand, fMLF, in the regulation of platelet function, hemostasis, and thrombosis is largely unknown. Fpr1-deficient mice and selective inhibitors for FPR1 were used to investigate the function of fMLF and FPR1 in platelets. N-formyl-methionyl-leucyl-phenylalanine primes platelet activation and augments thrombus formation, mainly through FPR1 in platelets. Formyl peptide receptor 1 plays a pivotal role in the regulation of platelet function. BACKGROUND: Formyl peptide receptors (FPRs) play pivotal roles in the regulation of innate immunity and host defense. The FPRs include three family members: FPR1, FPR2/ALX, and FPR3. The activation of FPR1 by its high-affinity ligand, N-formyl-methionyl-leucyl-phenylalanine (fMLF) (a bacterial chemoattractant peptide), triggers intracellular signaling in immune cells such as neutrophils and exacerbates inflammatory responses to accelerate the clearance of microbial infection. Notably, fMLF has been demonstrated to induce intracellular calcium mobilization and chemotaxis in platelets that are known to play significant roles in the regulation of innate immunity and inflammatory responses. Despite a plethora of research focused on the roles of FPR1 and its ligands such as fMLF on the modulation of immune responses, their impact on the regulation of hemostasis and thrombosis remains unexplored. OBJECTIVE: To determine the effects of fMLF on the modulation of platelet reactivity, hemostasis, and thrombus formation. METHODS: Selective inhibitors for FPR1 and Fpr1-deficient mice were used to determine the effects of fMLF and FPR1 on platelets using various platelet functional assays. RESULTS: N-formyl-methionyl-leucyl-phenylalanine primes platelet activation through inducing distinctive functions and enhances thrombus formation under arterial flow conditions. Moreover, FPR1 regulates normal platelet function as its deficiency in mouse or blockade in human platelets using a pharmacological inhibitor resulted in diminished agonist-induced platelet activation. CONCLUSION: Since FPR1 plays critical roles in numerous disease conditions, its influence on the modulation of platelet activation and thrombus formation may provide insights into the mechanisms that control platelet-mediated complications under diverse pathological settings.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , N-Formilmetionina Leucil-Fenilalanina , Ativação Plaquetária , Receptores de Formil Peptídeo/sangue , Trombose/induzido quimicamente , Animais , AMP Cíclico/sangue , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Transdução de Sinais , Trombose/sangue
5.
J Alzheimers Dis ; 67(1): 169-179, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30475772

RESUMO

Alzheimer's disease (AD) is characterized by progressive loss of memory and other cognitive functions. Accumulation of amyloid-ß (Aß) and hyperphosphorylated tau are two major neuropathological features of AD. Formyl peptide receptor 2 (FPR2), contributing to innate immunity and inflammation, has been implicated in the uptake and clearance of Aß. It remains unclear whether FPR2 affects cognition and tau phosphorylation. The effects of FPR2 in cognition and tau phosphorylation were examined using FPR2 knock-out (Fpr2-/-) mice receiving intracerebroventricular (ICV) injection of streptozotocin (STZ). The general behaviors and cognitive functions were evaluated using rotarod, open field test, and Morris water maze test. The alteration in tau hyperphosphorylation and activation of astrocytes were determined by using western blotting and/or immunofluorescence staining. ICV injection of STZ impaired spatial learning and memory of mice in Morris water maze. FPR2 deficiency improved spatial learning and memory of ICV-STZ mice. In the hippocampus and cortex of ICV-STZ mice, a marked increase was observed in tau phosphorylation at Ser199, Thr205, and Ser396 compared with ICV-saline control mice. However, FPR2 deficiency attenuated the hyperphosphorylation of tau at Ser199 and Ser396. In addition, the expression of GFAP was significantly increased in hippocampus and cortex of ICV-STZ mice. FPR2 deletion reduced the increase of GFAP expression induced by ICV injection of STZ. These results indicate that FPR2 deficiency is associate with improved cognition, reduced tau hyperphosphorylation, and activation of astrocytes in the mouse AD model tested. FPR2 may be a potential target in AD prevention and therapy.


Assuntos
Doença de Alzheimer/genética , Astrócitos/patologia , Cognição/fisiologia , Gliose/genética , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Proteínas tau/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Gliose/patologia , Injeções Intraventriculares , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Desempenho Psicomotor/efeitos dos fármacos , Estreptozocina/toxicidade
6.
FASEB J ; 32(1): 26-36, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28855276

RESUMO

Leukocytes express formyl-peptide receptors (FPRs), which sense microbe-associated molecular pattern (MAMP) molecules, leading to leukocyte chemotaxis and activation. We recently demonstrated that phenol-soluble modulin (PSM) peptides from highly pathogenic Staphylococcus aureus are efficient ligands for the human FPR2. How PSM detection by FPR2 impacts on the course of S. aureus infections has remained unknown. We characterized the specificity of mouse FPR2 (mFpr2) using a receptor-transfected cell line, homeobox b8 (Hoxb8), and primary neutrophils isolated from wild-type (WT) or mFpr2-/- mice. The influx of leukocytes into the peritoneum of WT and mFpr2-/- mice was analyzed. We demonstrate that mFpr2 is specifically activated by PSMs in mice, and they represent the first secreted pathogen-derived ligands for the mFpr2. Intraperitoneal infection with S. aureus led to lower numbers of immigrated leukocytes in mFpr2-/- compared with WT mice at 3 h after infection, and this difference was not observed when mice were infected with an S. aureus PSM mutant. Our data support the hypothesis that the mFpr2 is the functional homolog of the human FPR2 and that a mouse infection model represents a suitable model for analyzing the role of PSMs during infection. PSM recognition by mFpr2 shapes leukocyte influx in local infections, the typical infections caused by S. aureus-Weiss, E., Hanzelmann, D., Fehlhaber, B., Klos, A., von Loewenich, F. D., Liese, J., Peschel, A., Kretschmer, D. Formyl-peptide receptor 2 governs leukocyte influx in local Staphylococcus aureus infections.


Assuntos
Leucócitos/imunologia , Receptores de Formil Peptídeo/imunologia , Receptores de Lipoxinas/imunologia , Infecções Estafilocócicas/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Sinalização do Cálcio/imunologia , Degranulação Celular/imunologia , Linhagem Celular , Movimento Celular/imunologia , Modelos Animais de Doenças , Feminino , Genes Bacterianos , Proteínas de Homeodomínio/imunologia , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Neutrófilos/imunologia , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia
7.
J Immunol ; 198(4): 1718-1728, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28062700

RESUMO

Neutrophils are considered responsible for the pathophysiological changes resulting from hepatic ischemia-reperfusion (I/R) injury, which is a complication of trauma, shock, liver resection, and transplantation. Recently, evidence is accumulating that formyl-peptide receptor (FPR) signaling constitutes an important danger signal that guides neutrophils to sites of inflammation. This study aimed to investigate dynamic neutrophil recruitment using two-photon laser-scanning microscopy (TPLSM) in response to FPR1 blockade during hepatic I/R. LysM-eGFP mice were subjected to partial warm hepatic I/R. They were pretreated with an FPR1 antagonist, cyclosporine H (CsH), or formyl peptide, fMLF. Liver was imaged after hepatic laser irradiation or I/R using the TPLSM technique. CsH treatment alleviated hepatic I/R injury, as evidenced by decreased serum transaminase levels, reduced hepatocyte necrosis/apoptosis, and diminished inflammatory cytokine, chemokine, and oxidative stress. In contrast, systemic administration of fMLF showed few effects. Time-lapse TPLSM showed that FPR1 blockade inhibited the accumulation of neutrophils in the necrotic area induced by laser irradiation in vivo. In the CsH-treated I/R group, the number and crawling velocity of neutrophils in the nonperfused area were lower than those in the control group. Meanwhile, FPR1 blockade did not affect monocyte/macrophage recruitment. Hepatic I/R promoted the retention of neutrophils and their active behavior in the spleen, whereas CsH treatment prevented their changes. Intravital TPLSM revealed that formyl-peptide-FPR1 signaling is responsible for regulating neutrophil chemotaxis to allow migration into the necrotic area in hepatic I/R. Our findings suggest effective approaches for elucidating the mechanisms of immune cell responses in hepatic I/R.


Assuntos
Fígado/imunologia , Fígado/patologia , Infiltração de Neutrófilos , Receptores de Formil Peptídeo/metabolismo , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Apoptose , Quimiocinas/imunologia , Quimiotaxia de Leucócito , Ciclosporina/administração & dosagem , Citocinas/imunologia , Microscopia Intravital/métodos , Fígado/diagnóstico por imagem , Fígado/efeitos dos fármacos , Masculino , Camundongos , Monócitos/imunologia , Necrose , Neutrófilos/imunologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/deficiência , Traumatismo por Reperfusão/diagnóstico por imagem
8.
Sci Rep ; 6: 24244, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-27064029

RESUMO

Activation of the G-protein coupled formyl peptide receptor 2 (ALX/FPR2) by the lipid mediators lipoxin A4 and resolvin D1 (RvD1) promotes resolution of inflammation. Our previous in vitro studies indicate that RvD1 activation of ALX/FPR2 resolves cytokine-mediated inflammatory responses in mammalian cells. However, the impact of ALX/FPR2 activation on salivary gland function in vivo is unknown. The objective of this study was to determine whether submandibular glands (SMG) from ALX/FPR2(-/-) mice display enhanced inflammatory responses to lipopolysaccharides (LPS) stimulation. For these studies, C57BL/6 and ALX/FPR2(-/-) mice at age 8-12-week-old were treated with LPS by i.p for 24 h. Salivary gland structure and function were analyzed by histopathological assessment, saliva flow rate, quantitative PCR, Western blot analyses and immunofluorescence. Our results showed the following events in the ALX/FPR2(-/-) mice treated with LPS: a) upregulated inflammatory cytokines and decreased M3R (Muscarinic Acetylcholine receptor M3) and AQP5 (Aquaporin 5) protein expression, b) decreased saliva secretion, c) increased apoptosis, d) alteration of tight junction and neuronal damage. Overall, our data suggest that the loss of ALX/FPR2 results in unresolved acute inflammation and SMG dysfunction (xerostomia) in response to LPS that is similar to human salivary gland dysfunction induced by bacterial infection.


Assuntos
Receptores de Formil Peptídeo/metabolismo , Glândula Submandibular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Aquaporina 5/genética , Aquaporina 5/metabolismo , Citocinas/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Inflamação/etiologia , Inflamação/metabolismo , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Lipopolissacarídeos/toxicidade , Lipoxinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Glândula Submandibular/efeitos dos fármacos , Glândula Submandibular/patologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Regulação para Cima/efeitos dos fármacos , Xerostomia/etiologia , Xerostomia/metabolismo
9.
FASEB J ; 29(7): 2930-42, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25818588

RESUMO

Hypothalamo-pituitary-adrenocortical dysfunction contributes to morbidity and mortality in a high proportion of patients with sepsis. Here, we provide new insights into the underlying adrenal pathology. Using a murine model of endotoxemia (LPS injection), we demonstrate that adrenal insufficiency is triggered early in the disease. LPS induced a local inflammatory response in the adrenal gland within 4 hours of administration, coupled with increased expression of mRNAs for annexin A1 (AnxA1) and the formyl peptide receptors [(Fprs) 1, 2, and 3], a loss of lipid droplets in cortical cells (index of availability of cholesterol, the substrate for steroidogenesis), and a failure to mount a steroidogenic response to ACTH. Deletion of AnxA1 or Fpr2/3 in mice prevented lipid droplet loss, but not leukocyte infiltration. LPS increased adrenal myeloid differentiation primary response gene 88 and TLR2 mRNA expression, but not lymphocyte antigen 96 or TLR4. By contrast, neutrophil depletion prevented leukocyte infiltration and increased AnxA1, Fpr1, and Fpr3 mRNAs but had no impact on lipid droplet loss. Our novel data demonstrate that AnxA1 and Fpr2 have a critical role in the manifestation of adrenal insufficiency in this model, through regulation of cholesterol ester storage, suggesting that pharmacologic interventions targeting the AnxA1/FPR/ALX pathway may provide a new approach for the maintenance of adrenal steroidogenesis in sepsis.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Anexina A1/deficiência , Lipopolissacarídeos/toxicidade , Receptores de Formil Peptídeo/deficiência , Córtex Suprarrenal/patologia , Insuficiência Adrenal/induzido quimicamente , Insuficiência Adrenal/etiologia , Insuficiência Adrenal/metabolismo , Animais , Anexina A1/genética , Anexina A1/metabolismo , Ésteres do Colesterol , Corticosterona/biossíntese , Citocinas/sangue , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais
11.
Cardiovasc Res ; 105(1): 65-74, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25341894

RESUMO

AIMS: The formyl peptide receptor (FPR) subtype FPR2/ALX transduces pro-inflammatory responses and participates in the resolution of inflammation depending on activation. The aim of the present study was to unravel the role of FPR2/ALX signalling in atherosclerosis. METHODS AND RESULTS: Expression of FPR2/ALX was analysed in 127 human carotid atherosclerotic lesions and revealed that this receptor was expressed on macrophages, smooth muscle cells (SMCs), and endothelial cells. Furthermore, FPR2/ALX mRNA levels were significantly up-regulated in atherosclerotic lesions compared with healthy vessels. In multiple regression, age, creatinine, and clinical signs of increased cerebral ischaemia were independent predictors of FPR2/ALX expression. To provide mechanistic insights into these observations, we generated Ldlr(-/-)xFpr2(-/-) mice, which exhibited delayed atherosclerosis development and less macrophage infiltration compared with Ldlr(-/-)xFpr2(+/+) mice. These findings were reproduced by transplantation of Fpr2(-/-) bone marrow into Ldlr(-/-) mice and further extended by in vitro experiments, demonstrating a lower inflammatory state in Fpr2(-/-) macrophages. FPR2/ALX expression correlated with chemo- and cytokines in human atherosclerotic lesions and leucocytes. Finally, atherosclerotic lesions in Ldlr(-/-)xFpr2(-/-) mice exhibited decreased collagen content, and Fpr2(-/-) SMCs exhibited a profile of increased collagenase and decreased collagen production pathways. CONCLUSION: FPR2/ALX is proatherogenic due to effects on bone marrow-derived cells, but promoted a more stable plaque phenotype through effects on SMCs. Taken together, these results suggest a dual role of FPR2/ALX signalling in atherosclerosis by way of promoting disease progression and but increasing plaque stability.


Assuntos
Aterosclerose/etiologia , Aterosclerose/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Aterosclerose/genética , Transplante de Medula Óssea , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/metabolismo , Estenose das Carótidas/patologia , Colágeno/metabolismo , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores de Lipoxinas/genética , Transdução de Sinais , Regulação para Cima
12.
Circ Res ; 116(5): 827-35, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25520364

RESUMO

RATIONALE: Chemokine-controlled arterial leukocyte recruitment is a crucial process in atherosclerosis. Formyl peptide receptor 2 (FPR2) is a chemoattractant receptor that recognizes proinflammatory and proresolving ligands. The contribution of FPR2 and its proresolving ligand annexin A1 to atherosclerotic lesion formation is largely undefined. OBJECTIVE: Because of the ambivalence of FPR2 ligands, we here investigate the role of FPR2 and its resolving ligand annexin A1 in atherogenesis. METHODS AND RESULTS: Deletion of FPR2 or its ligand annexin A1 enhances atherosclerotic lesion formation, arterial myeloid cell adhesion, and recruitment. Mechanistically, we identify annexin A1 as an endogenous inhibitor of integrin activation evoked by the chemokines CCL5, CCL2, and CXCL1. Specifically, the annexin A1 fragment Ac2-26 counteracts conformational activation and clustering of integrins on myeloid cells evoked by CCL5, CCL2, and CXCL1 through inhibiting activation of the small GTPase Rap1. In vivo administration of Ac2-26 largely diminishes arterial recruitment of myeloid cells in a FPR2-dependent fashion. This effect is also observed in the presence of selective antagonists to CCR5, CCR2, or CXCR2, whereas Ac2-26 was without effect when all 3 chemokine receptors were antagonized simultaneously. Finally, repeated treatment with Ac2-26 reduces atherosclerotic lesion sizes and lesional macrophage accumulation. CONCLUSIONS: Instructing the annexin A1-FPR2 axis harbors a novel approach to target arterial leukocyte recruitment. With the ability of Ac2-26 to counteract integrin activation exerted by various chemokines, delivery of Ac2-26 may be superior in inhibition of arterial leukocyte recruitment when compared with blocking individual chemokine receptors.


Assuntos
Anexina A1/fisiologia , Doenças da Aorta/etiologia , Aterosclerose/etiologia , Animais , Anexina A1/deficiência , Anexina A1/genética , Anexina A1/farmacologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Quimiocina CCL2/fisiologia , Quimiocina CCL5/fisiologia , Quimiocina CXCL1/fisiologia , Quimiotaxia/efeitos dos fármacos , Gorduras na Dieta/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/fisiologia , Peptídeos/farmacologia , Receptores CCR2/antagonistas & inibidores , Receptores CCR5/fisiologia , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/fisiologia , Receptores de Interleucina-8B/antagonistas & inibidores , Proteínas rap1 de Ligação ao GTP/fisiologia
13.
PLoS One ; 9(12): e114626, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25517119

RESUMO

Formyl peptide receptors (FPR) belong to a family of sensors of the immune system that detect microbe-associated molecules and inform various cellular and sensorial mechanisms to the presence of pathogens in the host. Here we demonstrate that Fpr2/3-deficient mice show a distinct profile of behaviour characterised by reduced anxiety in the marble burying and light-dark box paradigms, increased exploratory behaviour in an open-field, together with superior performance on a novel object recognition test. Pharmacological blockade with a formyl peptide receptor antagonist, Boc2, in wild type mice reproduced most of the behavioural changes observed in the Fpr2/3(-/-) mice, including a significant improvement in novel object discrimination and reduced anxiety in a light/dark shuttle test. These effects were associated with reduced FPR signalling in the gut as shown by the significant reduction in the levels of p-p38. Collectively, these findings suggest that homeostatic FPR signalling exerts a modulatory effect on anxiety-like behaviours. These findings thus suggest that therapies targeting FPRs may be a novel approach to ameliorate behavioural abnormalities present in neuropsychiatric disorders at the cognitive-emotional interface.


Assuntos
Ansiedade/complicações , Ansiedade/tratamento farmacológico , Terapia de Alvo Molecular , Doenças do Sistema Nervoso/complicações , Doenças do Sistema Nervoso/tratamento farmacológico , Receptores de Formil Peptídeo/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/patologia , Corticosterona/sangue , Comportamento Exploratório/efeitos dos fármacos , Deleção de Genes , Masculino , Camundongos , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Reconhecimento Psicológico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
14.
Immunology ; 143(3): 447-61, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24863484

RESUMO

Bacterial meningitis is, despite progress in research and the development of new treatment strategies, still a cause of severe neuronal sequelae. The brain is protected from penetrating pathogens by both the blood-brain barrier and the innate immune system. The invading pathogens are recognized by pattern recognition receptors including the G-protein coupled formyl peptide receptors (FPRs), which are expressed by immune cells of the central nervous system. The expression of FPRs is up-regulated during bacterial meningitis, but the consequence on the progression of inflammation and impact on mortality are far from clear. Therefore, we used mFPR1 and mFPR2-deficient mice to investigate the effects on inflammation, bacterial growth and mortality in a mouse model of pneumococcal meningitis. Our results revealed increased bacterial burden, increased neutrophil infiltration and higher mortality in mFPR1/2-deficient mice in comparison to wild-type mice. The mFPR1- or mFPR2-deficient mice also showed significantly increased glial cell density, whereas the immune responses including the expression of anti-inflammatory cytokines and antimicrobial peptides were decreased in bacterial meningitis. Taken together, the results suggest that FPR1 and FPR2 play an important role in the innate immune responses against Streptococcus pneumoniae within the central nervous system and the lack of the receptors leads to a dysregulation of the inflammatory response compared with wild-type mice.


Assuntos
Meningite Pneumocócica/genética , Meningite Pneumocócica/mortalidade , Receptores de Formil Peptídeo/genética , Animais , Carga Bacteriana , Biomarcadores , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Contagem de Células , Sobrevivência Celular , Quimiocinas/genética , Modelos Animais de Doenças , Expressão Gênica , Imunidade Inata , Masculino , Meningite Pneumocócica/imunologia , Meningite Pneumocócica/microbiologia , Meningite Pneumocócica/patologia , Camundongos , Camundongos Knockout , Neuroglia/imunologia , Neuroglia/metabolismo , Neuroglia/patologia , Infiltração de Neutrófilos/imunologia , Receptores de Formil Peptídeo/deficiência , Índice de Gravidade de Doença
15.
Cell Host Microbe ; 14(3): 318-28, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24034617

RESUMO

Shifts in commensal microbiota composition are emerging as a hallmark of gastrointestinal inflammation. In particular, outgrowth of γ-proteobacteria has been linked to the etiology of inflammatory bowel disease and the pathologic consequences of infections. Here we show that following acute Toxoplasma gondii gastrointestinal infection of mice, control of commensal outgrowth is a highly coordinated process involving both the host response and microbial signals. Notably, neutrophil emigration to the intestinal lumen results in the generation of organized intraluminal structures that encapsulate commensals and limit their contact with the epithelium. Formation of these luminal casts depends on the high-affinity N-formyl peptide receptor, Fpr1. Consequently, after infection, mice deficient in Fpr1 display increased microbial translocation, poor commensal containment, and increased mortality. Altogether, our study describes a mechanism by which the host rapidly contains commensal pathobiont outgrowth during infection. Further, these results reveal Fpr1 as a major mediator of host commensal interaction during dysbiosis.


Assuntos
Disbiose , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/parasitologia , Neutrófilos/imunologia , Proteobactérias/imunologia , Receptores de Formil Peptídeo/metabolismo , Toxoplasma/crescimento & desenvolvimento , Animais , Carga Bacteriana , Translocação Bacteriana/imunologia , Modelos Animais de Doenças , Trato Gastrointestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteobactérias/crescimento & desenvolvimento , Receptores de Formil Peptídeo/deficiência , Análise de Sobrevida , Toxoplasmose Animal/parasitologia
16.
Sci Rep ; 2: 786, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139859

RESUMO

Listeria monocytogenes (Listeria) causes opportunistic infection in immunocompromised hosts with high mortality. Resistance to Listeria depends on immune responses and recruitment of neutrophils of the immune system into infected sites is an early and critical step. Mouse neutrophils express two G protein-coupled formylpeptide receptor subtypes Fpr1 and Fpr2 that recognize bacterial and host-derived chemotactic molecules including Listeria peptides for cell migration and activation. Here we report deficiency in Fprs exacerbated the severity of the infection and increased the mortality of infected mice. The mechanism involved impaired early neutrophil recruitment to the liver with Fpr1 and Fpr2 being sole receptors for neutrophils to sense Listeria chemoattractant signals and for production of bactericidal superoxide. Thus, Fprs are essential sentinels to guide the first wave of neutrophil infiltration in the liver of Listeria-infected mice for effective elimination of the invading pathogen.


Assuntos
Listeria monocytogenes/patogenicidade , Neutrófilos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Animais , Movimento Celular , Quimiocinas/metabolismo , Quimiotaxia , Células HEK293 , Humanos , Peróxido de Hidrogênio/metabolismo , Listeria monocytogenes/imunologia , Listeriose/microbiologia , Listeriose/mortalidade , Listeriose/patologia , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Camundongos Knockout , Infiltração de Neutrófilos , Neutrófilos/imunologia , Fagocitose , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética
17.
Dev Cell ; 22(5): 1079-91, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22542839

RESUMO

Neutrophil recruitment to inflammation sites purportedly depends on sequential waves of chemoattractants. Current models propose that leukotriene B(4) (LTB(4)), a secondary chemoattractant secreted by neutrophils in response to primary chemoattractants such as formyl peptides, is important in initiating the inflammation process. In this study we demonstrate that LTB(4) plays a central role in neutrophil activation and migration to formyl peptides. We show that LTB(4) production dramatically amplifies formyl peptide-mediated neutrophil polarization and chemotaxis by regulating specific signaling pathways acting upstream of actin polymerization and MyoII phosphorylation. Importantly, by analyzing the migration of neutrophils isolated from wild-type mice and mice lacking the formyl peptide receptor 1, we demonstrate that LTB(4) acts as a signal to relay information from cell to cell over long distances. Together, our findings imply that LTB(4) is a signal-relay molecule that exquisitely regulates neutrophil chemotaxis to formyl peptides, which are produced at the core of inflammation sites.


Assuntos
Polaridade Celular/fisiologia , Quimiotaxia de Leucócito/imunologia , Leucotrieno B4/metabolismo , Neutrófilos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Actinas/metabolismo , Animais , Comunicação Celular/fisiologia , Fatores Quimiotáticos/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Miosina Tipo II/metabolismo , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de Formil Peptídeo/deficiência , Transdução de Sinais
18.
Dig Dis Sci ; 57(7): 1802-12, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22383080

RESUMO

BACKGROUND: The receptor for formylated peptides, formyl peptide receptor 1 (FPR1), potently activates and serves as a chemoattractant receptor for neutrophils. AIM: Given the abundance of neutrophils in the inflamed colon, our aim was to determine if the FPR1 mediates colonic neutrophil migration, using the dextran sodium sulfate (DDS)-induced model of colitis. METHODS: Formyl peptide receptor 1 gene-deficient mice were administered DDS in drinking water for a single 5-day period (acute) or in two 5-day periods separated by 16 days (chronic). At the end of the treatment their colons were excised, measured, and prepared for histological evaluation. RESULTS: FPR1(-/-) mice experienced less severe acute colonic pathology than C57BL/6 (wildtype) mice. The opposite was observed following the second colitis cycle, with FPR1(-/-) mice developing worse pathology than wildtype mice. Both strains had similar numbers of infiltrating neutrophils in ulcerated areas of the colon after a single DSS cycle, but FPR1(-/-) mice had significantly more neutrophils in the ulcerated mucosa after two cycles. There was no difference in the capacity of neutrophils from each strain to migrate to chemoattractants. Since the FPR1(-/-) mice had larger ulcers compared to the wildtype mice, we propose that the FPR1(-/-) mice failed to recover at the same rate as wildtype mice. This apparent difference in restitution could not be attributed to observable differences in annexin A1. CONCLUSIONS: We conclude that neutrophil migration into the inflamed mouse colon does not depend on FPR1 but that FPR1 contributes in other pathological mechanisms that are harmful during acute inflammation but protective during chronic inflammation.


Assuntos
Colite/metabolismo , Colite/patologia , Colo/patologia , Suscetibilidade a Doenças/metabolismo , Neutrófilos/patologia , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/metabolismo , Doença Aguda , Animais , Anexina A1/metabolismo , Movimento Celular , Doença Crônica , Colite/induzido quimicamente , Doenças do Colo/epidemiologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Feminino , Incidência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Formil Peptídeo/genética , Úlcera/epidemiologia
19.
Nat Rev Immunol ; 9(1): 62-70, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19104500

RESUMO

Glucocorticoids are widely used for the management of inflammatory diseases. Their clinical application stems from our understanding of the inhibitory effect of the corticosteroid hormone cortisol on several components of the immune system. Endogenous and exogenous glucocorticoids mediate their multiple anti-inflammatory effects through many effector molecules. In this Opinion article, we focus on the role of one such effector molecule, annexin A1, and summarize the recent studies that provide insight into its molecular and pharmacological functions in immune responses. In addition, we propose a model in which glucocorticoids regulate the expression and function of annexin A1 in opposing ways in innate and adaptive immune cells to mediate the resolution of inflammation.


Assuntos
Anexina A1/fisiologia , Glucocorticoides/fisiologia , Sistema Imunitário/fisiologia , Inflamação/fisiopatologia , Animais , Anexina A1/deficiência , Anexina A1/genética , Anti-Inflamatórios/uso terapêutico , Ritmo Circadiano , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Hidrocortisona/fisiologia , Sistema Imunitário/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Lipoxinas/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Modelos Imunológicos , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/fisiologia , Receptores de Lipoxinas/fisiologia , Subpopulações de Linfócitos T/imunologia
20.
J Immunol ; 181(2): 1429-37, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606697

RESUMO

The prototypic formyl peptide N-formyl-Met-Leu-Phe (fMLF) is a major chemoattractant found in Escherichia coli culture supernatants and a potent agonist at human formyl peptide receptor (FPR) 1. Consistent with this, fMLF induces bactericidal functions in human neutrophils at nanomolar concentrations. However, it is a much less potent agonist for mouse FPR (mFPR) 1 and mouse neutrophils, requiring micromolar concentrations for cell activation. To determine whether other bacteria produce more potent agonists for mFPR1, we examined formyl peptides from Listeria monocytogenes and Staphylococcus aureus for their abilities to activate mouse neutrophils. A pentapeptide (N-formyl-Met-Ile-Val-Ile-Leu (fMIVIL)) from L. monocytogenes and a tetrapeptide (N-formyl-Met-Ile-Phe-Leu (fMIFL)) from S. aureus were found to induce mouse neutrophil chemotaxis at 1-10 nM and superoxide production at 10-100 nM, similar to the potency of fMLF on human neutrophils. Using transfected cell lines expressing mFPR1 and mFPR2, which are major forms of FPRs in mouse neutrophils, we found that mFPR1 is responsible for the high potency of fMIVIL and fMIFL. In comparison, activation of mFPR2 requires micromolar concentrations of the two peptides. Genetic deletion of mfpr1 resulted in abrogation of neutrophil superoxide production and degranulation in response to fMIVIL and fMIFL, further demonstrating that mFPR1 is the primary receptor for detection of these formyl peptides. In conclusion, the formyl peptides from L. monocytogenes and S. aureus are approximately 100-fold more potent than fMLF in activating mouse neutrophils. The ability of mFPR1 to detect bacterially derived formyl peptides indicates that this important host defense mechanism is conserved in mice.


Assuntos
Fatores Quimiotáticos/imunologia , Quimiotaxia de Leucócito , Listeria monocytogenes/imunologia , Neutrófilos/imunologia , Oligopeptídeos/imunologia , Receptores de Formil Peptídeo/metabolismo , Staphylococcus aureus/imunologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Formilmetionina Leucil-Fenilalanina/imunologia , Ativação de Neutrófilo , Neutrófilos/metabolismo , Oligopeptídeos/metabolismo , Ratos , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/imunologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...